CA2659858A1 - Liposome treatment of viral infections - Google Patents

Liposome treatment of viral infections Download PDF

Info

Publication number
CA2659858A1
CA2659858A1 CA002659858A CA2659858A CA2659858A1 CA 2659858 A1 CA2659858 A1 CA 2659858A1 CA 002659858 A CA002659858 A CA 002659858A CA 2659858 A CA2659858 A CA 2659858A CA 2659858 A1 CA2659858 A1 CA 2659858A1
Authority
CA
Canada
Prior art keywords
liposome
lipids
composition
dnj
infection
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002659858A
Other languages
French (fr)
Inventor
Raymond A. Dwek
Norica Nichita-Branza
Stefana Petrescu
Stephanie Pollock
Pauline Rudd
Christopher Scanlan
Nicole Zitzmann
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Oxford
Original Assignee
United Therapeutics Corporation
Raymond A. Dwek
Norica Nichita-Branza
Stefana Petrescu
Stephanie Pollock
Pauline Rudd
Christopher Scanlan
Nicole Zitzmann
University Of Oxford
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by United Therapeutics Corporation, Raymond A. Dwek, Norica Nichita-Branza, Stefana Petrescu, Stephanie Pollock, Pauline Rudd, Christopher Scanlan, Nicole Zitzmann, University Of Oxford filed Critical United Therapeutics Corporation
Publication of CA2659858A1 publication Critical patent/CA2659858A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6425Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a receptor, e.g. CD4, a cell surface antigen, i.e. not a peptide ligand targeting the antigen, or a cell surface determinant, i.e. a part of the surface of a cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers

Abstract

One can treat a viral infection such as hepatitis B (HBV), hepatitis C (HCV), and bovine viral diarrhea virus (BVDV) infections via the delivery of pH sensitive liposomes directly into the endoplasmic reticulum (ER) membrane. Two exemplary liposome formulations are DOPE/CHEMS (DC liposomes) and DOPE/CHEMS/PEG-PE (DCPP liposomes). DC and DCPP liposomes can optimize the intracellular delivery of N-butyl deoxynojirimycin (NB-DNJ), and consequently increase the in vivo activity of this iminosugar several orders of magnitude, and could be used in combination with other therapeutic agents such as interferon and/or ribavirin. The optimized release of NB-DNJ directly into the ER can be also applied for the treatment of other viruses, for which NB-DNJ is known to be an effective antiviral, such as human immunodeficiency virus (HIV).

Description

LIPOSOME TREATMENT OF VIRAL INFECTIONS
RELATED APPLICATIONS

[0001] The present application claims priority to US provisional applications Nos.
60/834,797 filed August 2, 2006, to Dwek et al. and 60/846,344 filed September 22, 2006, to Dwek et al., which are both incorporated herein by reference in their entirety.

FIELD
[0002] The present application relates generally to methods and compositions for treatment of viral infections and, more specifically, to methods and compositions for treatment of viral infections utilizing liposomes.

BACKGROUND
[0003] Hepatitis B virus. Hepatitis B virus (HBV, HepB) is a causative agent of acute and chronic liver disease including liver fibrosis, cirrhosis, inflammatory liver disease, and hepatic cancer that can lead to death in some patients, see e.g.
Joklik, Wolfgang K., Virology, Third Edition, Appleton & Lange, Norwalk, Conn., 1988 (ISBN 0-8385-9462-X). Although effective vaccines are available, more than 280 million people worldwide, i.e. 5 % of the world's population, are still chronically infected with the virus, see e.g. Locamini, S. A., et. al., Antiviral Chemistry &
Chemotherapy (1996) 7(2):53-64. Such vaccines have no therapeutic value for those already infected with the virus. In Europe and North America, between 0.1 %
and 1 % of the population is infected. Estimates are that 15 % to 20 % of individuals who acquire the infection develop cirrhosis or another chronic disability from HBV
infection. Once liver cirrhosis is established, morbidity and mortality are substantial, with about a 5 year patient survival period, see e.g. Blume, H., E., et.al., Advanced Drug Delivery Reviews (1995) 17:321-331.
[0004] Hepatitis C virus. Approximately 170 million people worldwide, i.e. 3 %
of the world's population, see e.g. WHO, J. Viral. Hepat. 1999; 6: 35-47, and approximately 4 million people in the United States are infected with Hepatitis C

virus (HCV, HepC). About 80 % of individuals acutely infected with HCV become chronically infected. Hence, HCV is a major cause of chronic hepatitis. Once chronically infected, the virus is almost never cleared without treatment. In rare cases, HCV infection causes clinically acute disease and even liver failure.
Chronic HCV infection can vary dramatically between individuals, where some will have clinically insignificant or minimal liver disease and never develop complications and others will have clinically apparent chronic hepatitis and may go on to develop cirrhosis. About 20 % of individuals with HCV who do develop cirrhosis will develop end-stage liver disease and have an increased risk of developing primary liver cancer.
[0005] Antiviral drugs such as interferon, alone or in combination with ribavirin, are effective in up to 80 % of patients (Di Bisceglie, A. M, and Hoofnagle, J. H.
2002, Hepatology 36, S121-S127), but many patients do not tolerate this form of combination therapy.
[0006] Bovine viral diarrhea virus. Bovine viral diarrhea virus (BVDV) is distributed worldwide and is prevalent in most cattle populations. BVDV is also commonly used as tissue culture surrogate of HCV. There are two viral biotypes of BVDV: noncytopathic (ncp) and cytopathic (cp). Classification of viral biotype is based on cytopathic effect in cultured cells and is not related to virulence.
Ncp BVDV
is common in cattle, while the cp biotype is relatively rare and arises from the ncp strain after a specific mutational event occurs in the viral genome. Infection of cells with the cp BVDV strain in tissue culture is characterized by formation of clusters of apoptotic cells (plaques) on the cell monolayer, which can be easily monitored microscopically.
[0007] Human immunodeficiency virus. Human immunodeficiency virus (HIV) is the causative agent of acquired immune deficiency syndrome (AIDS) and related disorders. There are at least two distinct types of HIV: HIV-1 and HIV-2.
Further, a large amount of genetic heterogeneity exists within populations of each of these types.
Since the onset of the AIDS epidemic, some 20 million people have died and the estimate is that over 40 million are now living with HIV-1/AIDS, with 14 000 people infected daily worldwide.
[0008] Numerous antiviral therapeutic agents and diagnostic capabilities have been developed that, at least for those with access, have greatly improved both the quantity and quality of life. Most of these drugs interfere with viral proteins or processes such as reverse transcription and protease activity. Unfortunately, these treatments do not eliminate infection, the unwanted effects of many therapies are severe, and drug resistant strains of HIV exist for every type of antiviral currently in use.
[0009] N-butyldeoxynojirimycin(NB-DNJ) as a therapeutic agent. NB-DNJ, also known as N-butyl-1,5-dideoxy-1,5-imino-D-glucitol, inhibits processing by the ER
glucosidases I and II, and has been shown to be an effective antiviral by causing the misfolding and/or ER-retention of glycoproteins of human immunodeficiency virus (HIV) and hepatitis viruses, such as Hepatitis B virus, Hepatitis C virus, Bovine viral diarrhea, virus amongst others. Methods of synthesizing NB-DNJ and other N-substituted deoxynojirimycin derivatives are described, for example, in US
parents Nos. 5,622,972, 4,246,345, 4,266,025, 4,405,714 and 4,806,650. Antiviral effects of NB-DNJ are discussed, for example, in US patents Nos. 6,465,487; 6,545,021;
6,689,759; 6,809,083 for hepatitis viruses and US patent No. 4,849,430 for HIV
virus.
[0010] Glucosidase inhibitors, such as NB-DNJ, have been shown to be effective in the treatment of HBV infection in both cell culture and using a woodchuck animal model, see e.g. T. Block, X. Lu, A.S. Mehta, B.S. Blumberg, B. Tennant, M.
Ebling, B. Korba, D.M. Lansky, G.S. Jacob & R.A. Dwek, Nat Med. 1998 May;4(5):610-4.
NB-DNJ suppresses secretion of HBV particles and causes intracellular retention of HBV DNA.
[0011] NB-DNJ has been shown to be a strong antiviral against BVDV, a cell culture model for HCV, see e.g. Branza-Nichita N, Durantel D, Carrouee-Durantel S, Dwek RA, Zitzmann N., J Virol. 2001 Apr;75(8):3527-36; Durantel, D., et al, J.
Virol., 2001, 75, 8987-8998; N. Zitzmann, et al, PNAS, 1999, 96, 11878-11882.
Treatment with NB-DNJ leads to decreased infectivity of viral progeny, with less of an effect on the actual number of secreted viruses.
[0012] NB-DNJ has been shown to be antiviral against HIV; treatment leads to a relatively small effect on the number of virus particles released from HIV-infected cells, however, the amount of infectious virus released is greatly reduced, see e.g. P.B.

Fischer, M. Collin, et al (1995), J. Virol. 69(9) :5791-7; P.B. Fischer, G.B.
Karlsson, T. Butters, R. Dwek and F. Platt, J. Virol. 70 (1996a), pp. 7143-7152, P.B.
Fischer, G.B. Karlsson, R. Dwek and F. Platt,. J. Virol. 70 (1996b), pp. 7153-7160.
Clinical trials involving NB-DNJ were conducted in HIV-1 infected patients, and results demonstrated that concentrations necessary for antiviral activity were too high and resulted in serious side-effects in patients, see e.g. Fischl M.A., Resnick L., Coombs R., Kremer A.B., Pottage J.C. Jr, Fass R.J., Fife K.H., Powderly W.G., Collier A.C., Aspinall R.L., et. al., J. Acquir. Immune. Defic. Syndr. 1994 Feb;.7(2):139-47. No mutant HIV strain resistant to NB-DNJ treatment currently exists.
[0013] ER protein folding & glucosidases I and H. The antiviral effect demonstrated by glucosidase inhibition is thought to be a result of misfolding or retention of viral glycoproteins within the ER, primarily through blocking entry into the calnexin/calreticulin cycle. Following transfer of the triglucosylated oligosaccharide (G1c3Man9GlcNAc2) to an Asn-X-Ser/Thr consensus sequence in the growing polypeptide chain, it is necessary that the three a-linked glucose residues be released before further processing to the mature carbohydrate units can take place.
Moreover, the two outer glucose residues must be trimmed to allow entry into the calnexin/calreticulin cycle for proper folding, see e.g. Bergeron, J.J. et.
al., Trends Biochem. Sci., 1994, 19, 124-128; Peterson, J. R. et. al., Mol. Biol. Cell, 1995, 6, 1173-1184. The initial processing is affected by an ER-situated integral membrane enzyme with a lumenally-oriented catalytic domain (glucosidase I) that specifically cleaves the al-2linked glucose residue; this is followed by the action of glucosidase II, which releases both of the al-3 linked glucose components.
[0014] Liposomes. Liposomes can deliver water-soluble compounds directly inside the cell, bypassing cellular membranes that act as molecular barriers. The pH
sensitive liposome formulation can involve the combination of phopsphatidylethanolamine (PE), or its derivatives (e.g. DOPE), with compounds containing an acidic group, which can act as a stabilizer at neutral pH.
Cholesteryl hemisuccinate (CHEMS) can be a good stabilizing molecule as its cholesterol group confers higher stability to the PE-containing vesicles compared to other amphiphilic stabilizers in vivo. The in vivo efficacy of liposome-mediated delivery can depend strongly on interactions with serum components (opsonins) that can influence their pharmacokinetics and biodistribution. pH-sensitive liposomes can be rapidly cleared from blood circulation, accumulating in the liver and spleen, however inclusion of lipids with covalently attached polyethylene glycol (PEG) can overcome clearance by the reticuloendothelial system (RES) by stabilizing the net-negative charge on DOPE:CHEMS liposomes, leading to long circulation times. DOPE-CHEMS and DOPE-CHEMS-PEG-PE liposomes and methods of their preparation are described, for example, in V.A. Slepushkin, S. Simoes, P. Dazin, M.S. Newman, L.S. Guo and M.C.P. de Lima, J. Biol. Chem. 272 (1997) 2382-2388; and S. Simoes, V.
Slepushkin, N. Duzgunes and M.C. Pedroso de Lima, Biomembranes 1515 (2001) 23-37, both incorporated herein by reference in their entirety.
[0015] Delivery of NB-DNJ encapsulated in DOPE-CHEMS (molar ratio 6:4) is disclosed in US patent application No. US2003/0124160.

SUMMARY
[0016] One embodiment provides a method of treating a viral infection, comprising administering to a host in need thereof a composition comprising (a) a liposome comprising DOPE and CHEMS lipids and (b) one or more therapeutic agents encapsulated into the liposome, wherein the viral infection is an ER membrane budding viral infection or a plasma membrane budding viral infection; wherein the one or more therapeutic agents comprise N-butyl deoxynojirimycin (NB-DNJ) and wherein said administering results in delivering of the one or more therapeutic agents into an endoplasmic reticulum of a cell, that is infected with a virus causing the infection, and incorporating one or more lipids of the liposome in an endoplasmic reticulum membrane of the cell.
[0017] Another embodiment of the invention provides a method of treating a viral infection, comprising administering to a host in need thereof a composition comprising (a) a liposome comprising DOPE, CHEMS and PEG-PE lipids and (b) one or more therapeutic agents encapsulated into the liposome. The one or more therapeutic agents can comprise N-butyl deoxynojirimycin (NB-DNJ).
[0018] Yet another embodiment provides a method comprising administering to a host in need thereof a composition comprising (a) a pH sensitive liposome and (b) an antigen encapsulated inside the liposome, wherein the administering results in increasing antigen presentation by a major histocompatibility molecule class 1 of a antigen presenting cell.
[0019] Yet another embodiment is a method of treating an HIV infection comprising administering to a host in need thereof a composition comprising a liposome conjugated with a gp120/gp4l complex targeting moiety.
[0020] And yet another embodiment is a composition comprising a liposome comprising DOPE, CHEMS and PEG-PE lipids and at least one therapeutic agent, such as N-butyl deoxynojirimycin (NB-DNJ) encapsulated inside the liposome.
[0021] And yet another embodiment is a composition, comprising a pH sensitive liposome and an antigen encapsulated inside the liposome.
[0022] And yet another embodiment is a composition comprising a liposome conjugated with a gp120/gp4l complex targeting moiety.
[0023] And yet according to another embodiment, a method of treating or preventing a viral infection, comprises administering to a host in need thereof a composition comprises (a) a liposome comprising PI lipids and (b) at least one antiviral therapeutic agent encapsulated into the liposome, wherein said contacting results in delivering of the at least one therapeutic agent into the ER lumen of a cell, that is infected with a virus causing the infection, and incorporating one or more lipids of the liposome in the ER membrane of the cell.
[0024] And yet according to another embodiment, a composition comprises a liposome comprising PI lipids and at least one antiviral therapeutic agent encapsulated inside the liposome.
[0025] And yet another embodiment is a method of treating a viral infection comprising administering to a host in need thereof a composition comprising (a) a liposome comprising PI lipids and (b) at least one antiviral protein intercalated into a lipid layer of the liposome, wherein said contacting results in incorporating one or more lipids of the liposome in an endoplasmic reticulum membrane of a cell, that is infected with a virus causing the infection.
[0026] And yet another embodiment is a composition comprising (a) a liposome comprising PI lipids and (b) at least one antiviral protein intercalated into a lipid layer of the liposome.
[0027] And yet another embodiment is a composition comprising a liposome comprising PI lipids and at least one therapeutic agent encapsulated inside the liposome.
[0028] And yet another embodiment is a method of treating or preventing a physiological condition comprising administering to a subject in need thereof a composition comprising a liposome comprising PI lipids and at least one therapeutic agent encapsulated inside the liposome.
[0029] And yet another embodiment is a composition comprising a liposome comprising PI lipids and at least one protein intercalated into a lipid bilayer of the liposome.
[0030] And yet another embodiment is a method of treating or preventing a physiological condition comprising administering to a subject in need thereof a composition comprising a liposome comprising PI lipids and at least one protein intercalated into a lipid bilayer of the liposome.

BRIEF DESCRIPTION OF THE DRAWINGS
[0031] FIG. 1 demonstrates endoplasmic reticulum (ER) localization of dequenched calcein and fluorescence labelled lipids (Rh-PE) following delivery via DCPP-Rh liposomes.
[0032] FIG. 2 shows toxicity of DCPP liposomes in CHO and MDBK cells. DCPP
liposomes encapsulating PBS were added to CHO cells with final lipid concentrations ranging between 0 - 500 gM, and to MDBK cells with concentrations ranging between 0 - 150 gM. Cells and liposomes were left to incubate 5 days before cell viability was measured by trypan blue staining. Results are presented as the percentage of viable cells compared to the untreated control.
[0033] FIG. 3 is a plot showing DCPP-Rh liposome uptake and intracellular calcein dequenching in CHO cells with the encapsulation of deoxynojirimycin (DNJ), N-butyl deoxynojirimycin (NB-DNJ) and N-nonyl deoxynojirimycin (NN-DNJ) compounds.

DCPP-Rh liposomes were prepared encapsulating each compound at two different concentrations. Liposome uptake, measured by incorporation of Rh-PE in cellular membranes, and calcein dequenching, a measure of intracellular release, was determined following a 5-min pulse with liposomes and a 30-min chase in a fresh medium.
[0034] FIG. 4 shows pH sensitivity of DCPP liposomes containing various DNJ
molecules.
[0035] FIG. 5 is a plot showing BVDV secretion following treatment with 1VB-DNJ:
free vs. DCPP liposome-mediated delivery. The secretion of BVDV particles from infected MDBK cells during treatment with NB-DNJ added either freely into the medium or via liposomes with a final lipid concentration of 50 gM was determined by real-time PCR following a 3 day incubation. Results are presented as a percentage of RNA copies detected by real-time PCR compared to the untreated control.
[0036] FIG. 6 demonstrates the effects of NB-DNJ on ncp BVDV infectivity: free vs. DCPP liposome-mediated delivery. Infectivity of ncp BVDV particles produced by infected MDBK cells in the presence of 1VB-DNJ, either added freely in the medium or via liposomes with a final lipid concentration of 50 gM, was measured by incubation with naive MDBK cells for 3 days. Infected cells were detected by immunofluorescent staining of non-structural BVDV proteins present in MDBK
cells using DAPI counterstain as a control. Data from BVDV secretion (Figure 2) were used to normalize calculations for final percent infectivity.
[0037] FIG. 7 shows the antiviral effect of NB-DNJ against cp BVDV: free vs.
DC
liposome-mediated delivery. MDBK cells infected with cp BVDV were grown in the presence of free or DC liposome-included NB-DNJ, for 3 days. The supematants containing secreted virus were used to infect naive MDBK. After 3 days the resulting plaques were counted under the microscope (yield assay).
[0038] FIG. 8 demonstrates inhibition of glycan processing on the HIV envelope protein gp120 expressed in the presence of NB-DNJ: free vs. DCPP liposome-mediated delivery. CHO cells expressing a soluble form of gp120 were incubated with NB-DNJ, either added freely into the medium or via liposomes with a final lipid concentration of 100 gM. NB-DNJ activity, determined by the inhibition of glucose trimming by ER glucosidases, was measured by the binding of monoclonal antibodies 2G12 and bl2 to the NB-DNJ-treated gp120 in capture ELISAs.
[0039] FIG. 9 (A-C) presents antiviral effects of free NB-DNJ on eight separate HIV-1 primary isolates. FIG. 9A shows average p24 secretion of the eight isolates treated with varying concentrations of NB-DNJ over four weeks. Error bars show the standard deviation between isolates for each treatment. FIG. 9B shows secretion of each primary isolate following initial treatment with free NB-DNJ at concentrations ranging 0 - 1 mM. FIG. 9C shows sensitivity of individual primary isolates to NB-DNJ following three weeks of treatment. All values are expressed as a percentage of the untreated control, and data represent the mean obtained from triplicate samples of two independent experiments. The approximate IC50s and IC90s for each treatment are indicated by grey (dotted) lines across the graph.
[0040] FIG. 10 A-H represent a data demonstrating how liposomes increase the antiviral activity of NB-DNJ against eight primary isolates of HIV-1. PBMCs infected with each isolate (represented in graphs A to H) were treated with liposomes (L) encapsulating NB-DNJ at various concentrations over a four week period.
Treatment with 500 M NB-DNJ free (F) in the media is shown as a reference for antiviral activity. The legend indicates the final concentration of NB-DNJ for each treatment. All values are expressed as a percentage of the untreated control, and data represent the mean obtained from triplicate samples of two independent experiments.
[0041] FIG. 11 shows uptake of sCD4-liposomes and immunoliposomes by cells infected with a broad range of HIV-1 isolates. sCD4- and MAb-liposome conjugates are incubated with PBMCs infected with nine different primary isolates (clades in brackets). Increased uptake is measured by the increase in fluorescent lipids in cells following incubation. All values were normalized to the `no infection, liposome only' control and data represent the mean standard error obtained from triplicates of one experiment. The relative uptake data were tested separately for each infection using a series of one-way analyses of variance followed by post-hoc Tukey tests to test for differences between the effectiveness of different targeting molecules.
Significant differences in uptake between liposome conjugates and the liposome only control (P<0.0001) are denoted with asterisks.
[0042] FIG. 12 A-H demonstrate potent synergistic antiviral activity of sCD4-liposomes encapsulating NB-DNJ. PBMCs infected with each isolate (represented in graphs A to H) were treated with sCD4-liposomes (CD4-L) encapsulating NB-DNJ
at various concentrations over a four week period. Treatment with 500 M NB-DNJ
free (F) in the media is shown as a reference for antiviral activity. The legend indicates the final concentration of NB-DNJ for each treatment. All values are expressed as a percentage of the untreated control, and data represent the mean obtained from triplicate samples of one experiment.
[0043] FIG. 13 A-E show representative fluorescent images of rhodamine labelled PE inside MDBK cells following a 15 m pulse with various liposome preparations. In particular, FIG. 13A demonstrates data for DOPE:CHEMS:Rh-PE (6:4:0.1); FIG.

for DOPC:CHEMS:Rh-PE (6:4:0.1); FIG. 13C for DOPE:CHEMS:PI:Rh-PE
(6:4:1:0.1); FIG. 13D DOPE:CHEMS:PI:Rh-PE (6:4:2:0.1) and FIG. 13E
DOPE: CHEMS:PI:Rh-PE (6:4:3:0.1). Intracellular localization of Rh-PE was observed for each liposome preparation at time points: 0, 1, 2, 5, 24 and 48 hours.
DAPI counterstain was used to visualize all cells [0044] FIG. 14 shows results from treating both cp-BVDV-infected and uninfected MDBK cells with various liposome preparations containing Rh-labelled PE and measuring the secretion of the Rh-PE lipid following three days incubation. An increase in the Rh-PE secretion between infected and uninfected cells treated with the same liposome composition was due to secretion of viral particles, which have budded from the ER membrane, containing the Rh-PE lipid.

DETAILED DESCRIPTION
[0045] Unless otherwise specified, "a" or "an" means "one or more."
Definition of terms:
[0046] As used herein, the term "viral infection" describes a diseased state, in which a virus invades a healthy cell, uses the cell's reproductive machinery to multiply or replicate and ultimately lyse the cell resulting in cell death, release of viral particles and the infection of other cells by the newly produced progeny viruses. Latent infection by certain viruses is also a possible result of viral infection.
[0047] As used herein, the term "treating or preventing viral infection" means to inhibit the replication of the particular virus, to inhibit viral transmission, or to prevent the virus from establishing itself in its host, and to ameliorate or alleviate the symptoms of the disease caused by the viral infection. The treatment is considered therapeutic if there is a reduction in viral load, decrease in mortality and/or morbidity.
[0048] The term "therapeutic agent" refers to any agent, such as a molecule or a compound, which can assist in treating a physiological condition, such as a viral infection or a disease caused thereby.
[0049] The term "synergistic" as used herein refers to a combination, which is more effective than the additive effects of any two or more single therapeutic agents. A
synergistic effect as used herein refers to the ability to use lower amounts (doses) of either single therapy to treat or prevent a physiological condition, such as a viral infection or a disease caused thereby. The lower doses can result in lower toxicity without reducing efficacy of the treatment. In addition, a synergistic effect can result in improved efficacy, e.g. in an improved antiviral activity. Finally, for a viral infection, synergy may result in an improved avoidance or reduction of a viral resistance against a single therapeutic agent or single therapy.
[0050] Liposomes can be defined as organic compounds comprising lipids in a spherical bilayer formation. Liposomes discussed herein may include one or more lipids represented by the following abbreviations:
CHEMS stands for cholesteryl hemisuccinate lipid.
DOPE stands for dioleoylphosphatidylethanolamine lipid.
DOPC stands for dioleoylphosphatidylcholine lipid.
PE stands for phosphatidylethanolamine lipid.
PEG-PE stands for polyethylene glycol (2000)-distearoylphosphatidylethanolamine lipid.
Rh-PE stands for lissamine rhodamine B-phosphatidylethanolamine lipid.
MCC-PE stands for 1,2-Dioleoyl-sn-Glycero-3-Phosphoethanolamine-N-[4-(p-maleimidomethyl)cyclohexane-carboxamide] lipid.
PI stands for phosphatidylinositol lipid.

The term "intracellular delivery" refers to the delivery of encapsulated material from liposomes into any intracellular compartment.
IC50 or IC90 (inhibitory concentration 50 or 90) is a concentration of a therapeutic agent used to achieve 50% or 90% reduction of viral infection, respectively.
A DC liposome designates a liposome comprising DOPE and CHEMS lipids with a molar ratio of 6:3.
A DCPP liposome designates a liposome comprising DOPE, CHEMS and PEG-PE
lipids with a molar ratio of 6:4:0.3.
PBMC stands for peripheral blood mononuclear cell.
sCD4 stands for a soluble CD4 molecule. By "soluble CD4" or "sCD4" or D I D2"
is meant a CD4 molecule, or a fragment thereof, that is in aqueous solution and that can mimic the activity of native membrane-anchored CD4 by altering the conformation of HIV Env, as is understood by those of ordinary skill in the art. One example of a soluble CD4 is the two-domain soluble CD4 (sCD4 or DID2) described, e.g., in Salzwedel et al. J. Virol. 74:326 333, 2000.
MAb stands for a monoclonal antibody.
DNJ denotes deoxynojirimycin.
NB-DNJ denotes N-butyl deoxynojirimycin.
NN-DNJ denotes N-nonyl deoxynojirimycin.
BVDV stands for bovine viral diarrhea virus.
HBV stands for hepatitis B virus.
HCV stands for hepatitis C virus.
HIV stands for human immunodeficiency virus.
Ncp stands for non-cytopatic.
Cp stands for cytopatic.
ER stands for endoplasmic reticulum.
CHO stands for Chinese hamster ovary cells MDBK stands for Madin-Darby bovine kidney cells.
PCR stands for polymerase chain reaction.
FOS stands for free oligosaccharides.
HPLC stands for high performance liquid chromatography.

PHA stands for phytohemagglutinin.
FBS stands for fetal bovine serum.
TCID50 stands for 50% tissue culture infective dose.
ELISA stands for Enzyme Linked Immunosorbent Assay.
IgG stands for immunoglobuline.

DAPI stands for 4',6-Diamidino-2-phenylindole.
PBS stands for phosphate buffered saline.

LIPOSOMES TREATMENT OF VIRAL INFECTIONS
[0051] The inventors have discovered that, upon contacting a cell, a pH
sensitive liposome, that comprises DOPE, CHEMS and/or PEG-PE lipids, such as a DC
liposome or a DCPP liposome, can be able to bypass the cell's endosomal pathway following endocytosis and deliver a material encapsulated in the liposome directly into the cell's endoplasmic reticulum (ER), i.e. in the ER lumen. One or more lipids of the liposome can also integrate into the ER membrane of the cell. This discovery can have a major implication for the treatment of viral infections, for which the virus requires budding from the ER membrane, such as HBV, HCV and BVDV infections, as incorporation of liposome lipids with the ER membrane of a cell infected with the virus can alter the envelope of budding virus particles and, thus, reduce infectivity.
[0052] The inventors have also discovered that encapsulation of N-butyl deoxynojirimycin (NB-DNJ) in a DCPP liposome can provide an increased intracellular delivery as compared to DCPP encapsulation of other deoxynojirimycin compounds, such as deoxynojirimycin (DNJ) or N-nonyl deoxynojirimycin (NN-DNJ). Such an increased intracellular delivery of NB-DNJ can lead to an enhancement of in vivo activity of NB-DNJ.
[0053] Furthermore, the inventors have discovered that a liposome comprising DOPE, CHEMS and/or PEG-PE lipids, such as a DCPP liposome, can have an antiviral effect of its own, i.e. independent of any therapeutic agents encapsulated inside the liposome, and that the DCPP liposome and a therapeutic agent, such as NB-DNJ, that is encapsulated inside the liposome can act synergistically against the virus.
[0054] Accordingly, one embodiment is a method of treating an ER membrane virus budding infection, i.e. a viral infection, for which virus budding can occur at the ER
membrane, such as HBV, HCV or BVDV infection. The method can involve contacting a cell infected with a virus responsible for the infection, with a composition, in which NB-DNJ is encapsulated in a liposome that comprises DOPE
and CHEMS lipids. Such contacting can provide a synergistic therapy resulting in delivering one or more lipids of the liposome to the ER membrane of the contacted cell, altering the membrane and thereby reducing infectivity of progeny viruses, and by releasing the encapsulated NB-DNJ directly into the cell's ER lumen.
[0055] Another embodiment is a method of treating a viral infection by contacting a cell, that is infected with a virus responsible for the infection, with a composition that contains 1) a liposome comprising DOPE, CHEMS and PEG-PE lipids and 2) a therapeutic agent that is encapsulated inside the liposome. The viral infection can be, for example, HCV, HBV, BVDV, HIV, Moloney murine leukaemia virus, murine hepatitis virus, herpes simplex virus types 1 and 2, cytomegalovirus, Sindbis virus, Semliki forest virus, Vesicular stomatis virus, Influenza A virus, Measles virus, Dengue virus, or Japanese Encephalitis virus, as described in R. A. Dwek, et al, Nat.
Rev. Drug Discov. 2002 Jan; 1(1):65-75.
[0056] In some embodiments, the therapeutic agent encapsulated inside the liposome can be, an a-glucosidase inhibitor. In some embodiments, the a-glucosidase inhibitor can be ER a-glucosidase inhibitor. In general, any virus that relies on interactions with calnexin and/or calreticulin for proper folding of its viral envelope glycoproteins, can be targeted with ER a-glucosidase inhibitor.
[0057] The alpha-glucosidase inhibitor can be an agent that inhibits host alpha-glucosidase enzymatic activity by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90%, or more, compared to the enzymatic activity of the alpha-glucosidase in the absence of the agent. The term "alpha-glucosidase inhibitot"
encompasses both naturally occuring and synthetic agents that inhibit host alpha-glucosidase activity.
[0058] Suitable alpha-glucosidase inhibitors include, but not limited to, deoxynojirimycin and N-substituted deoxynojirimycins, such as compounds of Formula II and pharmaceutically acceptable salts thereof:

OX
WO///,/,, "``oOY

OZ
N
I

II
, [0059] where Ri is selected from substituted or unsubstituted alkyl groups, substituted or unsubstituted cycloalkyl groups, substituted or unsubstituted aryl groups, or substituted or unsubstituted oxaalkyl groups, selected from but not limited to arylalkyl, cycloalkylalkyl, branched or straight chain alkyl groups, and oxaalkyl groups; and where W, X, Y, and Z are each independently selected from hydrogen, alkanoyl groups, aroyl groups, and haloalkanoyl groups.
[0060] In some of such embodiments, Rl is selected from ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, neopentyl, isopentyl, hexyl, -(CH2)20(CH2)5CH3, -(CH2)20(CH2)6CH3, -(CH2)6OCH2CH3, and -(CH2)2OCH2CH2CH3. In other such embodiments, Rl is butyl, and W, X, Y, and Z are all hydrogen.
[0061] In some embodiments, the compound of Formula II is selected from, but is not limited to N-(n-hexyl-)-1,5-dideoxy-1,5-imino-D-glucitol; N-(n-heptyl-)-1,5-dideoxy-1,5-imino-D-glucitol; N-(n-octyl-)-1,5-dideoxy-1,5-imino-D-glucitol; N-(n-octyl-)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(n-nonyl-)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(n-decyl-)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(n-undecyl-)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate;
N-(n-nonyl-)-1,5-dideoxy-1,5-imino-D-glucitol; N-(n-decyl-)-1,5-dideoxy-1,5-imino-D-glucitol; N-(n-undecyl-)-1,5-dideoxy-1,5-imino-D-glucitol; N-(n-dodecyl-)-1,5-dideoxy-1,5-imino-D-glucitol; N-(2-ethylhexyl)-1,5-dideoxy-1,5-imino-D-glucitol;
N-(4-ethylhexyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(5-methylhexyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(3-propylhexyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(1-pentylpentylhexyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(1-butylbutyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(7-methyloctyl-)-1,5-dideoxy-1,5-imino-D-glucitol; N-(8-methylnonyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(9-methyldecyl)-1,5-dideoxy-l,5-imino-D-glucitol; N-(10-methylundecyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(6-cyclohexylhexyl-)-1,5-dideoxy-1,5-imino-D-glucitol; N-(4-cyclohexylbutyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(2-cyclohexylethyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(1-cyclohexylmethyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(1-phenylmethyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(3-phenylpropyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(3-(4-methyl)-phenylpropyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(6-phenylhexyl)-1,5-dideoxy-1,5-imino-D-glucitol; N-(n-nonyl-)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(n-decyl-)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(n-undecyl-)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(n-dodecyl-)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate;
N-(2-ethylhexyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(4-ethylhexyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(5-methylhexyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(3-propylhexyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(1-pentylpentylhexyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(1-butylbutyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(7-methyloctyl-)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(8-methylnonyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(9-methyldecyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(10-methylundecyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(6-cyclohexylhexyl-)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(4-cyclohexylbutyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate;
N-(2-cyclohexylethyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(1-cyclohexylmethyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(1-phenylmethyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(3-phenylpropyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(3-(4-methyl)-phenylpropyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; N-(6-phenylhexyl)-1,5-dideoxy-1,5-imino-D-glucitol, tetrabutyrate; pharmaceutically acceptable salts thereof;
and mixtures of any two or more thereof.
[0062] Suitable alpha-glucosidase inhibitors also include N-oxaalkylated deoxynojirimycins such as N-hydroxyethyl DNJ (Miglitol or Glyset ) described in US patent 4,639,436.
[0063] Suitable alpha-glucosidase inhibitors also include castanospermines and castanospermine derivatives, such as compounds of Formula (I) and pharmaceutically acceptable salts thereof disclosed in US patent application No. 2006/0194835, including 6-0-butanoyl castanospermine (celgosivir), and compounds and pharmaceutically acceptable salt thereof of Formula II disclosed in PCT
publication No. W001054692.
[0064] In some embodiments, the alpha glucosidase inhibitor can be acarbose (O-4,6-dideoxy-4-[[(1S,4R,5S,6S)-4,5,6-trihydroxy-3-(hydroxymethyl)-2-cyc-lohexen-1-yl]amino]-a-D-glucopyranosyl-(1--->4)-O---->-D-gluc- opyranosyl-(1--->4)-D-glucose), or Precose . Acarbose is disclosed in U.S. Pat. No. 4,904,769. In some embodiments, the alpha glucosidase inhibitor can be a highly purified form of acarbose (see, e.g., U.S. Pat. No. 4,904,769).
[0065] In some embodiments, the therapeutic agent encapsulated inside the liposome can be an ion channel inhibitor. In some embodiments, the ion channel inhibitor can be an agent inhibiting the activity of HCV p7 protein. Ion channel inhibitors and methods of identifying them are detailed in US patent publication 2004/0110795. Suitable ion channel inhibitors include compounds of Formula I
and pharmaceutically acceptable salts thereof, including N-(7-oxa-nonyl)-1,5,6-trideoxy-1,5-imino-D-galactitol (N-7-oxa-nonyl 6-MeDGJ or UT231B) and N-10-oxaundecul-6-MeDGJ. Suitable ion channel inhibitors also include, but not limited to, N-nonyl deoxynojirimycin, N-nonyl deoxynogalactonojirimycin and N-oxanonyl deoxynogalactonojirimycin.
[0066] In some embodiments, the therapeutic agent encapsulated inside the liposome can include an iminosugar. Suitable iminosugars include both naturally occurring iminosugars and synthetic iminosugars.
[0067] In some embodiments, the iminosugar can be deoxynojirimycin or N-substituted deoxynojirimycin derivative. Examples of suitable N-substituted deoxynojirimycin derivatives include, but not limited to, compounds of Formula II of the present application, compounds of Formula I of US patent No. 6,545,021 and N-oxaalkylated deoxynojirimycins, such as N-hydroxyethyl DNJ (Miglitol or Glyset ) described in US patent 4,639,436.
[0068] In some embodiments, the iminosugar can be castanospermine or castanospermine derivative. Suitable castanospemine derivatives include, but not limited to, compounds of Formula (I) and pharmaceutically acceptable salts thereof disclosed in US patent application No. 2006/0194835 and compounds and pharmaceutically acceptable salt thereof of Formula II disclosed in PCT
publication No. W001054692.
[0069] In some embodiments, the iminosugar can be deoxynogalactojirimycin or N-substituted derivative thereof such as those disclosed in PCT publications No.
W099/24401 and WO01/10429. Examples of suitable N-substituted deoxynogalactojirimycin derivatives include, but not limited to, N-alkylated deoxynogalactojirimycins (N-alkyl-1,5-dideoxy-1,5-imino-D-galactitols), such as N-nonyl deoxynogalactojirimycin, and N-oxa-alkylated deoxynogalactojirimycins (N-oxa-alkyl-1,5-dideoxy-1,5-imino-D-galactitols), such as N-7-oxanonyl deoxynogalactojirimycin.
[0070] In some embodiments, the iminosugar can be N-substituted 1,5,6-trideoxy-1,5-imino-D-galactitol (N-substituted MeDGJ) including, but not limited to compounds of Formula I:

OH
HO/4, OH

I
[0071] wherein R is selected from substituted or unsubstituted alkyl groups, substituted or unsubstituted cycloalkyl groups, substituted or unsubstituted heterocyclyl groups, or substituted or unsubstituted oxaalkyl groups. In some embodiments, substituted or unsubstituted alkyl groups and/or substituted or unsubstituted oxaalkyl groups comprise from 1 to 16 carbon atoms, or from 4 to carbon atoms or from 8 to 10 carbon atoms. In some embodiments, substituted or unsubstituted alkyl groups and/or substituted or unsubstituted oxaalkyl groups comprise from 1 to 4 oxygen atoms, and from 1 to 2 oxygen atoms in other embodiments. In other embodiments, substituted or unsubstituted alkyl groups and/or substituted or unsubstituted oxaalkyl groups comprise from 1 to 16 carbon atoms and from 1 to 4 oxygen atoms. Thus, in some embodiments, R is selected from, but is not limited to -(CHz)60CH3, -(CHz)6OCH2CH3, -(CH2)60(CH2)2CH3, -(CHZ)60(CHZ)3CH3, -(CHZ)20(CHZ)5CH3, -(CHZ)20(CHZ)6CH3, and -(CH2)20(CH2)7CH3. N-substituted MeDGJs are disclosed, for example, in PCT
publication No. WO01/10429.
In some embodiments, the therapeutic agent encapsulated inside the liposome can include a nitrogen containing compound having formula VIII or a pharmaceutically acceptable salt thereof:

I
R12 (VIII), wherein Ri2 is an alkyl such as C1-C20, or C1-C6 or C7-C12 or C8-C16 and can also contain from 1 to 5 or from 1 to 3 or from 1 to 2 oxygen, R'2 can be an oxa-substituted alkyl derivative. Examples if oxa-substituted alkyl derivatives include 3-oxanonyl, 3-oxadecyl, 7-oxanonyl and 7-oxadecyl.

R2 is hydrogen, R3 is carboxy, or a C1-C4 alkoxycarbonyl, or R~ and R3, together X y \ /

are-(C)n- or -(CXY)n-, wherein n is 3 or 4, each X, independently, is hydrogen, hydroxy, amino, carboxy, a C1-C4 alkylcarboxy, a C1-C4 alkyl, a C1-C4 alkoxy, a C1-C4 hydroxyalkyl, a C1-C6 acyloxy, or an aroyloxy, and each Y, independently, is hydrogen, hydroxy, amino, carboxy, a C1-C4 alkylcarboxy, a C1-C4 alkyl, a C1-alkoxy, a C1-C4 hydroxyalkyl, a C1-C6 acyloxy, an aroyloxy, or deleted (i.e.
not present);

R4 is hydrogen or deleted (i.e. not present); and R5 is hydrogen, hydroxy, amino, a substituted amino, carboxy, an alkoxycarbonyl, an aminocarbonyl, an alkyl, an aryl, an aralkyl, an alkoxy, a hydroxyalkyl, an acyloxy, or an aroyloxy, or R3 and R5, together, form a phenyl and R4 is deleted (i.e. not present).
In some embodiments, the nitrogen containing compound has the formula:

Y X
X Y X Y
Y X Y X

N N

> >
X

X X
X \ X

I I I
~
R5 N(D X R5 N X

R$

H H
i-H R1102C I
R12 , or R12 where each of R6-R10, independently, is selected from the group consisting of hydrogen, hydroxy, amino, carboxy, C1-C4 alkylcarboxy, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 hydroxyalkyl, C1-C4 acyloxy, and aroyloxy; and Rii is hydrogen or C1-C6 alkyl.
The nitrogen-containing compound can be N-alkylated piperidine, N-oxa-alkylated piperidine, N-alkylated pyrrolidine, N-oxa-alkylated pyrrolidine, N-alkylated phenylamine, N-oxa-alkylated phenylamine, N-alkylated pyridine, N-oxa-alkylated pyridine, N-alkylated pyrrole, N-oxa-alkylated pyrrole, N-alkylated amino acid, or N-oxa-alkylated amino acid. In certain embodiments, the N-alkylated piperidine, N-oxa-alkylated piperidine, N-alkylated pyrrolidine, or N-oxa-alkylated pyrrolidine compound can be an iminosugar. For example, in some embodiments, the nitrogen-containing compound can be N-alkyl-1,5-dideoxy-1,5-imino-D-galactitol (N-alkyl-DGJ) or N-oxa-alkyl-1,5-dideoxy-1,5-imino-D-galactitol (N-oxa-alkyl-DGJ) having the formula:

OH
H O H
~~~=.

or N-alkyl- 1,5,6-trideoxy- 1,5 -imino-D-galactitol (N-alkyl-MeDGJ) or N-oxa-alkyl-1,5,6-trideoxy-1,5-imino-D-galactitol having (N-oxa-alkyl-MeDGJ) having the formula:

OH
HOH
[0072] As used herein, the groups have the following characteristics, unless the number of carbon atoms is specified otherwise. Alkyl groups have from 1 to 20 carbon atoms and are linear or branched, substituted or unsubstituted. Alkoxy groups have from 1 to 16 carbon atoms, and are linear or branched, substituted or unsubstituted. Alkoxycarbonyl groups are ester groups having from 2 to 16 carbon atoms. Alkenyloxy groups have from 2 to 16 carbon atoms, from 1 to 6 double bonds, and are linear or branched, substituted or unsubstituted. Alkynyloxy groups have from 2 to 16 carbon atoms, from 1 to 3 triple bonds, and are linear or branched, substituted or unsubstituted. Aryl groups have from 6 to 14 carbon atoms (e.g., phenyl groups) and are substituted or unsubstituted. Aralkyloxy (e.g., benzyloxy) and aroyloxy (e.g., benzoyloxy) groups have from 7 to 15 carbon atoms and are substituted or unsubstituted. Amino groups can be primary, secondary, tertiary, or quatemary amino groups (i.e., substituted amino groups). Aminocarbonyl groups are amido groups (e.g., substituted amido groups) having from 1 to 32 carbon atoms.
Substituted groups can include a substituent selected from the group consisting of halogen, hydroxy, C1_io alkyl, Cz_io alkenyl, C1_io acyl, or C1_10 alkoxy.
[0073] The N-alkylated amino acid can be an N-alkylated naturally occurring amino acid, such as an N-alkylated a-amino acid. A naturally occurring amino acid is one of the 20 common a-amino acids (Gly, Ala, Val, Leu, Ile, Ser, Thr, Asp, Asn, Lys, Glu, Gln, Arg, His, Phe, Cys, Trp, Tyr, Met, and Pro), and other amino acids that are natural products, such as norleucine, ethylglycine, omithine, methylbutenyl-methylthreonine, and phenylglycine. Examples of amino acid side chains (e.g., R 5) include H (glycine), methyl (alanine), -CH2C(O)NH2 (asparagine), -CH2-SH
(cysteine), and -CH(OH)CH3 (threonine).
[0074] An N-alkylated compound can be prepared by reductive alkylation of an amino (or imino) compound. For example, the amino or imino compound can be exposed to an aldehyde, along with a reducing agent (e.g., sodium cyanoborohydride) to N-alkylate the amine. Similarly, a N-oxa-alkylated compound can be prepared by reductive alkylation of an amino (or imino) compound. For example, the amino or imino compound can be exposed to an oxa-aldehyde, along with a reducing agent (e.g., sodium cyanoborohydride) to N-oxa-alkylate the amine.
[0075] The nitrogen-containing compound can include one or more protecting groups. Various protecting groups are well known. In general, the species of protecting group is not critical, provided that it is stable to the conditions of any subsequent reaction(s) on other positions of the compound and can be removed at the appropriate point without adversely affecting the remainder of the molecule.
In addition, a protecting group may be substituted for another after substantive synthetic transformations are complete. Clearly, where a compound differs from a compound disclosed herein only in that one or more protecting groups of the disclosed compound has been substituted with a different protecting group, that compound is within the invention. Further examples and conditions are found in Greene, Protective Groups in Organic Chemistry, (lst Ed., 1981, Greene & Wuts, 2"d Ed., 1991).
[0076] The nitrogen-containing compound can be purified, for example, by crystallization or chromatographic methods. The compound can be prepared stereospecifically using a stereospecific amino or imino compound as a starting material.
[0077] The amino and imino compounds used as starting materials in the preparation of the long chain N-alkylated compounds are commercially available (Sigma, St. Louis, MO; Cambridge Research Biochemicals, Norwich, Cheshire, United Kingdom; Toronto Research Chemicals, Ontario, Canada) or can be prepared by known synthetic methods. For example, the compounds can be N-alkylated imino sugar compounds or oxa-substituted derivatives thereof. The imino sugar can be, for example, deoxygalactonojirmycin (DGJ), 1-methyl-deoxygalactonojirimycin (MeDGJ), deoxynorjirimycin (DNJ), altrostatin, 2R,5R-dihydroxymethyl-3R,4R-dihydroxypyrrolidine (DMDP), or derivatives, enantiomers, or stereoisomers thereof.
[0078] The syntheses of a variety of iminosugar compounds have been described.
For example, methods of synthesizing DNJ derivatives are known and are described, for example, in U.S. Patent Nos. 5,622,972, 5,401,645, 5,200,523, 5,043,273, 4,994,572, 4,246,345, 4,266,025, 4,405,714, and 4,806,650. Methods of synthesizing other iminosugar derivatives are known and are described, for example, in U.S.
Patent Nos. 4,861,892, 4,894,388, 4,910,310, 4,996,329, 5,011,929, 5,013,842, 5,017,704, 5,580,884, 5,286,877, and 5,100,797 and PCT publication No. WO 01/10429. The enantiospecific synthesis of 2R,5R-dihydroxymethyl-3R,4R-dihydroxypyrrolidine (DMDP) is described by Fleet & Smith (Tetrahedron Lett. 26:1469-1472, 1985).
[0079] The contacted cell can be a cell from a mammal, such as a human. In some cases, contacting the infected cell with the liposome composition can be done through administering the composition to a subject that comprises the infected cell.
The subject can be a mammal, such as a human. In some embodiments, the liposomal composition can be administered by intravenous injection. Yet in some embodiments, the liposomal composition can be administered via a parenteral routes other than intravenous injection, such as intraperitoneal, subcutaneous, intradermal, intraepidermal, intramuscular or transdermal route. Yet in some embodiments, the liposomal composition can be administered via a mucosal surface, e.g. an ocular, intranasal, pulmonary, intestinal, rectal and urinary tract surfaces.
Administration routes for liposomal compositions are disclosed, for example, in A. S. Ulrich, Biophysical Aspects of Using Liposomes as Delivery Vehicles, Bioscience Reports, Volume 22, Issue 2, Apr 2002, 129 - 150.
[0080] Delivery of a therapeutic agent, such as NB-DNJ, via the liposome into the ER lumen can lower an effective amount of the therapeutic agent required for inhibition of ER-glucosidase compared to non-liposome methods. For example, for NB-DNJ, the IC90 can be reduced by at least 100, or by at least 500, or by at least 1000, or by at least 5000, or by at least 10000, or by at least 50000 or by at least 100000. Such a reduction of the effective antiviral amount of NB-DNJ can result in final concentrations of administered NB-DNJ that are one or more orders of magnitude below toxic levels in mammals, in particular, humans.
[0081] In some cases, the liposome composition comprising a therapeutic agent, such as NB-DNJ, can be contacted with the infected cell in combination with one or more additional therapeutic agents, such as antiviral agents. In some cases, such additional therapeutic agents can be co-encapsulated with NB-DNJ into the liposome.
Yet in some cases, contacting the infected cell with such additional therapeutic agents can be a result of administering the additional therapeutic agents to a subject comprising the cell. The administration of the additional therapeutic agents can be carried out by adding the therapeutic agents to the composition. Yet in some cases, the administration of the additional therapeutic agents can be performed separately from administering the liposome composition containing NB-DNJ. Such separate administration can be performed via an administration pathway that can the same or different that the administration pathway used for the liposome composition.
[0082] Combination therapy may not only reduce the effective dose of an agent required for antiviral activity, thereby reducing its toxicity, but may also improve the absolute antiviral effect as a result of attacking the virus through multiple mechanisms. For example, lipids of the DCPP liposome and 1VB-DNJ can act 1) at an envelope of the virus, where treatment with NB-DNJ containing liposomes can alter the envelope's composition with the addition of foreign lipids and 2) through misfolding of viral glycoproteins, thereby reducing the infectivity. Thus, the liposome encapsulating 1VB-DNJ used in combination with one or more agents, that has targets or mechanisms of action different from NB-DNJ, can provide additive or synergistic effect.
[0083] In addition, combination therapy can provide means for circumventing or decreasing a chance of development of viral resistance.
[0084] The particular additional therapeutic agent(s) that can be used in combination the liposome containing NB-DNJ can depend of the viral infection being treated. For example, for a hepatitis infection, such as HBV, HCV or BVDV
infection, such therapeutic agent(s) can be a nucleoside or nucleotide antiviral agent and/or an immunostimulating/ immunomodulating agent. Various nucleoside agents, nucleotide agents and immunostimulating/ immunomodulating agents that can be used in combination with 1VB-DNJ for treatment of hepatitis are exemplified in US
patent No. 6,689,759 issued February 10, 2004, to Jacob et. al. For example, for treatment of hepatitis C infection, NB-DNJ can be encapsulated in the liposome in combination with 1-b-D-ribofuranosyl-lH-1,2,4-triazole-3-carboxamide (ribavirin), as a nucleoside agent, and interferon such as interferon alpha, as an immunostimulating/ immunomodulating agent. The treatment of hepatitis infections with ribavirin and/or interferon is discussed, for example, in US patents Nos.
6,172,046; 6,177,074; 6,299,872; 6,387,365; 6,472,373; 6,524,570 and 6,824,768.
[0085] For treating an HIV infection, a therapeutic agent that can be used in combination with a liposome containing NB-DNJ can be an anti-HIV agent, which can be, for example, nucleoside Reverse Transcriptase (RT) inhibitor, such as (-)-2'-deoxy-3'-thiocytidine-5'-triphosphate (3TC); (-)-cis-5-fluoro-l-[2-(hydroxy-methyl)-[ 1,3 -oxathiolan-5 -yl] cyto sine (FTC); 3'-azido-3'-deoxythymidine (AZT) and dideoxy-inosine (ddl); a non-nucleoside RT inhibitors, such as Nl 1-cyclopropyl-4-methyl-5,11-dihydro-6H-dipyrido[3,2-b:2'3'-e]-[1,4] diazepin-6-one (Neviparine), a protease inhibitor or a combination thereof. Anti HIV therapeutic agents can be used in double or triple combinations, such as AZT, DDI, and nevirapin combination.

LIPOSOMES CONJUGATED WITH gp120/gp41 TARGETTING MOIETY
[0086] The invention also provides a composition comprising a liposome conjugated with a gp120/gp4l targeting moiety and a method of treating an HIV
infection by contacting an cell infected with the infection with such composition. The gp120/gp4l targeting moiety can comprise a sCD4 molecule or a monoclonal antibody, such as IgG 2F5 or IgG b12 antibodies. In some embodiments, the liposome can comprise DOPE and CHEMS lipids. In some embodiments, the liposome can further comprise PEG-PE lipids. In some embodiments, the liposome can further comprise MCC-PE lipids. For treatment of the HIV infection, the composition can further comprise an additional therapeutic agent, such as NB-DNJ, encapsulated inside the liposome.

LIPOSOMES COMPRISING PI LIPIDS
[0087] The inventors have also discovered that a liposome, such as a pH
sensitive liposome, that comprises phosphatidylinositol (PI) lipids, can target more effectively the ER membrane of a cell that a liposome that does not contain PI lipids.
Furthermore, the liposome comprising PI lipids can increase a lifetime in the cell of one or more lipids delivered via the liposome.
Accordingly, in some embodiments, the invention provides a composition that includes a liposome comprising PI lipids and at least one therapeutic agent, such as an antiviral therapeutic agent, encapsulated inside the liposome and a method for targeted delivery comprising administering such a composition to a subject, which can be a mammal, such as a human. Such a targeted delivery method can be used for treating or preventing a physiological condition, such as a viral infection or a disease caused thereof, in a subject affected by the condition.
[0088] Inventors further believe that incorporation of the at least one antiviral protein into ER-targeting liposome may synergistically reduce viral infectivity due to 1) direct delivery of lipids of the liposome into the ER membrane that when incorporated into viral envelope can reduce viral infectivity and 2) direct delivery of the at least one antiviral protein into the ER membrane that can incorporate into the viral envelope of budding particles and independently reduce infectivity. The encapsulation of at least one therapeutic agent, such as a antiviral therapeutic agent, into the liposome can provide additional synergistic effect due to 3) direct delivery of the therapeutic agent into intracellular compartments, such as ER lumen.
[0089] Accordingly, in some embodiments, the invention provides a composition that includes a liposome comprising PI lipids and at least one protein, such as an antiviral protein, intercalated into a lipid bilayer of the liposome and a method for targeted delivery comprising administering such a composition to a subject, which can be a mammal, such as a human. Such a targeted delivery method can be used for treating or preventing a physiological condition, such as a viral infection or a disease caused thereof, in a subject affected by the condition.
[0090] Yet in some embodiments, the invention provides a composition that includes a liposome comprising PI lipids, at least one therapeutic agent, such as an antiviral composition, encapsulated inside the liposome and at least one protein, such as an antiviral protein, intercalated into a lipid bilayer of the liposome and a method for targeted delivery comprising administering such a composition to a subject, which can be a mammal, such as a human.
[0091] The viral infection can be, for example, an ER membrane budding viral infection, i.e. a viral infection, for which virus budding occurs at the ER
membrane, such as HBV, HCV or BVDV infection, or a plasma membrane budding viral infection, i.e. a viral infection, for which virus budding occurs at the plasma membrane, such as an HIV infection.
[0092] The liposome comprising PI lipids can further contain one or more lipids such as DOPE, CHEMS and/or PEG-PE lipids. A molar concentration of PI lipids in the liposome can vary from about 3% to about 60% or from about 5% to about 50%
or from about 10% to about 30%.
[0093] In some embodiments, the therapeutic agent encapsulated inside the liposome comprising the PI lipids can be a-glucosidase inhibitor, such as any of a-glucosidase inhibitors discussed above.
[0094] In some embodiments, the therapeutic agent encapsulated inside the liposome comprising the PI lipids can be an ion channel activity inhibitor discussed above.
[0095] In some embodiments, the therapeutic agent encapsulated inside the liposome comprising the PI lipids can be an iminosugar, such as any of the iminosugars discussed above.
[0096] In some embodiments, the therapeutic agent encapsulated inside the liposome comprising the PI lipids can be a nitrogen containing compound of formula VIII.
[0097] In some embodiments, a protein intercalated into a liposome containing PI
lipids can be an antiviral protein. Suitable antiviral proteins include, but are not limited to, viral receptors known to bind viral envelope proteins and/or mutated forms of viral envelope proteins and/or proteins known to interfere with viral envelope interactions. For example, for an HIV infection, the antiviral protein can be a CD4 protein. For HCV, BVDV, or HBV infections, the antiviral protein can be a mutated version of one of their respective viral envelope proteins, such as El or E2 proteins for HCV/BVDV.
[0098] The invention is further illustrated by, though in no way limited to, the following examples.

LIPOSOME PREPARATION
[0099] Liposomes used in all experiments were prepared as follows: chloroform solutions of lipids were placed into glass tubes, and the solvent was evaporated under a stream of nitrogen followed by vacuum centrifugation under reduced pressure.

Lipid films (10 gmoles total lipid) were hydrated by vortexing in 1 ml PBS
buffer, pH
7.4 (with or without drug and/or with 80 mM calcein in PBS) for 1 h at room temperature. The resulting multilamellar vesicles were sonicated in a bath-type sonicator for 15 min followed by extrusion 21 times through a polycarbonate filter of 80-nm pore diameter.

OPTIMIZATION OF ENCAPSULANT CONCENTRATION
[0100] The fluorescent molecule, calcein, was encapsulated inside DCPP
liposomes at a concentration of 80 mM, at which concentration its fluorescence is self-quenched.
Leakage of calcein from the liposomes, and its dilution into the surrounding medium, results in dequenching and an increase in measurable/observable fluorescence.
Percent encapsulation was determined by diluting final liposome preparations containing calcein in MES-buffered saline, pH 7.4, to a final phospholipid concentration of 0.5 gM, and calcein was measured at ~,X = 490 and ~'m = 520 nm, before and after the addition of Triton X-100 to a final concentration of 0.1 %. The difference in fluorescence following the addition of detergent is taken as the percent encapsulation within liposomes, and is used to estimate the amount of DNJ-compounds to determine the final concentrations of compounds in each experiment.

LIPID ENCAPSULATION INTO THE ER MEMBRANE
[0101] Liposomes are able to deliver encapsulated material directly into the lumen of the ER while lipids are incorporated into the ER membrane. Initial evidence comes from tagging liposomes with a fluorescent label, where rhodamine conjugated to PE
(Rh-PE) was incorporated into DCPP liposomes (1 % of molar content), so that lipid content was DOPE:CHEMS:PEG-PE:Rh-PE (DCPP-Rh), molar ratio of 6:4:0.3:0.1.
MDBK cells were pulsed for 30 min with Rh-PE-labeled and calcein containing pH-sensitive liposomes. Unadsorbed liposomes were removed and the cells were further chased for 3 h. At the end of the chase period, cells were incubated for 30 min with ER-Tracker, an ER marker, washed and visualized. The merged picture shown in Figure 1 demonstrates that calcein, the Rh-labelled lipid and the ER tracker co-localize. Based on this, one can conclude that liposomes deliver their aqueous content to the ER, after an initial fusion step with the ER membrane shown by the co-localization of the liposomes lipids with the ER-tracker, a dye which integrates within the ER membrane.

LIPOSOME TOXICITY
[0102] The toxicity of liposomes are measured in two different cell lines:
Chinese hamster ovary (CHO) and Madin-Darby bovine kidney (MDBK) cells.
[0103] Cells were seeded in 6-well plates to over 80 % confluency, and DCPP
liposomes encapsulating PBS were added to the media with a final lipid concentration ranging 0 - 500 gM for CHO cells, and 0 - 150 gM for MDBK cells. Cells and liposomes were left to incubate at 37 C for 5 days before cells were harvested and counted following staining with trypan blue. Results are expressed as the percentage of viable cells present in treated samples as compared to the untreated control (=
100% on the x axis), and are the mean S.D. of duplicates from three separate experiments.
[0104] Results are demonstrated in Figure 2, where cytotoxicity in CHO cells gradually appeared following incubation in the presence of 150-gM lipid concentrations. MDBK cells appeared to be more sensitive to the DCPP
liposomes, and demonstrated severe cytotoxicity at lipid concentrations greater than 75 gM.

IMINOSUGAR RELEASE FROM DCPP LIPOSOMES
[0105] In all experiments with CHO and MDBK cells, DCPP liposomes have been added to a final lipid concentration of 100 gM and 50 gM, respectively (> 95%
cell viability for both).
[0106] In the following example, the butyl chain of NB-DNJ is shown to be directly responsible for the increased intracellular release of encapsulated material from DCPP
liposomes.
[0107] The effects of different DNJ compounds on the cellular uptake and intracellular release, when encapsulated inside DCPP liposomes, was determined by diluting compounds in 80-mM calcein and incorporating Rh-PE in liposome membranes as previously described, with the final liposome composition being DOPE:CHEMS:PEG-PE:Rh-PE (DCPP-Rh, 6:4:0.3:0.1) Liposomes encapsulating calcein alone, 10-gM DNJ, 1-mM DNJ, 10-gM NB-DNJ, 1-mM 1VB-DNJ, 10-gM
NN-DNJ, or 1-mM NN-DNJ, were incubated with CHO cells for 45 min before liposomes were removed and cells were washed twice in 1 x PBS. Cells were then analyzed in a fluorometer to measure calcein dequenching (~'X = 490 and ~'m =

nm) and rhodamine fluorescence (~,X = 584 and ~,m = 612 nm). Data represent the mean S.D. obtained from triplicates of four independent experiments, where mean rhodamine fluorescence values reflect the binding and uptake of liposomes and the mean calcein fluorescence reflects the intracellular dequenching of the dye (i.e. dye released from liposomes into intracellular compartments). Results are expressed as the percent fluorescence measured following incubation with DNJ-containing liposomes as compared to the calcein/PBS-only liposome control (= 100% on the x axis).
[0108] As shown in Figure 3, DNJ and NN-DNJ both had no effect on the amount of calcein dequenching, and, therefore, on intracellular release. NB-DNJ, however, demonstrates a concentration-dependent increase in dequenched calcein, where gM and 1-mM formulations result in approximate 1.8- and 2.3-fold increases, respectively.
[0109] The calculated ratio of calcein to rhodamine fluorescence is taken as a measure of the amount of aqueous marker released per cell-associated liposome, and represents the efficiency of intracellular delivery. Efficiencies calculated for each encapsulated material are listed in Table 1. Results demonstrate that 10 gM
and 1 mM NB-DNJ-containing liposomes increased the efficiency of intracellular delivery from DCPP liposomes by approximately 2- and 3-fold, respectively.
Table 1. Efficiency of intracellular release from DCPP liposomes determined by the fluorescent measurements of dequenched calcein and Rh-PE incorporation taken from Figure 4.
Encapsulated Material Efficiency (calcein/rhodamine, AU) calcein only 10.5 2.1 gM DNJ 10.4 1.8 1 mM DNJ 10.8 2.0 gM NB-DNJ 21.7 1.5 1 mM NB-DNJ 28.4 2.4 10 gM NN-DNJ 4.4 1.3 1 mM NN-DNJ 3.2 1.1 [0110] Although the present invention is not bound by its theory of operation, it is probable that a mechanism, by which the butyl chain of NB-DNJ promotes increased intracellular delivery of encapsulated material from DCPP liposomes, is through the destabilization of liposomes by insertion of the butyl chain within the lipid bilayer.
The nine-carbon alkyl chain of NN-DNJ can also insert itself into the liposomes' lipid bilayer; however, at this length the molecule may actually stabilize the bilayer formation, as suggested by the presented results. Although NN-DNJ liposomes lead to an increase in cellular uptake, the amount of calcein released remains comparable to controls indicating increased liposome stability within the cell.

STABILITY OF DCPP LIPOSOMES
[0111] In the following example the alkyl chains of both NB-DNJ and NN-DNJ are shown to change the properties of DCPP liposomes by making them more stable at lower pH.
[0112] DCPP liposomes encapsulating 1-mM DNJ, NB-DNJ, or NN-DNJ in 80-mM
calcein buffer were compared with empty (calcein only) liposomes to determine the effects of the alkyl chain of NB-DNJ and NN-DNJ on the pH-sensitivity. 10 g1 of calcein-loaded DCPP liposomes (final phospholipid concentration, 5 gM) was added to 2 ml of MES-buffered saline at various pH values ranging from 5.0 - 7.4, and left to incubate with shaking 15 min at 37 C. Following incubation, calcein fluorescence was measured before and after the addition of Triton to a final concentration of 0.1 %.
Fluorescence intensities obtained at acidic pH values were corrected for the slight effect of pH on calcein fluorescence. The percentage of calcein release at each pH

was calculated using the formula: % leakage =((Iõ-Io)/(I100-Io)) x 100, where Io is the fluorescence at neutral pH, Iõ is the corrected intensity at acidic pH before the addition of Triton, and I100 is the total dequenched calcein at neutral pH.
[0113] Results are demonstrated in Figure 4, where no difference in stability between calcein only and DNJ encapsulated liposomes was observed. A
significant decrease in pH sensitivity was observed, however, with NB-DNJ and NN-DNJ
liposomes, indicating increased stability at low pH as a result of the alkyl chains.
Although both NB-DNJ and NN-DNJ conferred greater stability to DCPP liposomes in vitro, only NN-DNJ actually inhibited calcein delivery in vivo (as observed in Figure 4 and Table 1), highlighting the presence of factors other than pH
sensitivity that influence or control the process of intracellular cargo release.
[0114] Based on the findings from Figure 3, Figure 4 and Table 1, one can conclude that not only the presence, but also the length of the alkyl chain on the DNJ
molecule can affect the properties of liposomes when encapsulated inside. These observations can be most likely a result of the alkyl chains inserting into the lipid bilayer of the liposomes, and effectively changing the lipid composition. Although only a 4 carbon chain (NB-DNJ) and a 9 carbon chain (NN-DNJ) have been used in these studies, it seems as though shorter chains can destabilize the liposomes in vivo leading to increased intracellular release of cargo molecules, whereas longer chains can stabilize the same lipid composition so that release is inhibited.

DECREASE OF BVDV SECRETION BY NB-DNJ ENCAPSULATED IN DCPP
LIPOSOMES
[0115] In the following example, NB-DNJ encapsulated in DCPP liposomes is shown to decrease the secretion of BVDV particles from MDBK cells.
[0116] The combined antiviral effects of NB-DNJ incorporated into DC and DCPP
liposomes have been shown in MDBK cells infected with either the cp or ncp strain of BVDV.
[0117] In a first study using the ncp strain of BVDV, NB-DNJ was added freely to the culture medium, so that final concentrations ranged between 0 and 750 nM.
In separate incubations, NB-DNJ DCPP liposomes were added so that the final lipid concentration was 50 gM and final concentrations of NB-DNJ ranged between 0 and 750 nM. Experiments were carried out in duplicate in 6-well plates. The quantity of secreted BVDV viral particles was measured following 3 days of incubation.
Virus secretion analysis was performed by quantitative PCR (real-time PCR) on viral RNA
extracted from 500 g1 of supematant. Real-time PCR used primers directed against the ncp BVDV RNA where the forward primer sequence was TAGGGCAAACCATCTGGAAG, and the reverse primer sequence was ACTTGGAGCTACAGGCCTCA. Results are expressed as the percentage of RNA
copies present in treated samples as compared to the untreated control (= 100%
on the x axis).
[0118] Figure 5 represents results on the effects of NB-DNJ, both free and in DCPP
liposomes, on the secretion of ncp BVDV. Using a final concentration of 750 nM
NB-DNJ, there is a 2-fold decrease in the number of secreted viral particles with liposome-mediated delivery, and no effect with free delivery. Assays can be further optimized to decrease viral secretion even further through increasing either liposome or encapsulated NB-DNJ concentrations.
[0119] Although the present invention is not limited by its theory of operation, the mechanism, by which BVDV secretion could be inhibited, may be through the retention of viral glycoproteins, such as E l and E2, within the ER as a result of glucosidase inhibition.

BVDV INFECTIVITY
[0120] In the following example, DCPP liposomes, both empty and encapsulating NB-DNJ, are shown to decrease the infectivity of BVDV particles secreted from MDBK cells. DCPP liposomes and NB-DNJ are also shown to work synergistically.
[0121] Using BVDV viral particles collected from the previous experiment (500 g1 of supematant containing secreted BVDV), the infectivity of secreted NB-DNJ-treated BVDV was determined by incubation with naive MDBK cells for 3 days followed by immunofluorescent staining to identify BVDV proteins in infected cells. Cells were stained post-infection using an anti-BVDV NS2-NS3 monoclonal antibody, followed by a FITC-labelled secondary antibody. Experiments were performed in duplicate using 6-well plates. The percent viral infectivity was calculated by the number of infected cells, identified by the presence of non-structural BVDV proteins, divided by the total cell count determined by DAPI staining of cell nuclei. Results were also normalized to account for decreased or increased viral titres in each sample (results from BVDV secretion experiments).
[0122] Figure 6 represents results on the effects of NB-DNJ, both free and in DCPP
liposomes, on the infectivity of ncp BVDV. These results demonstrate that untreated BVDV, as well as BVDV treated with free NB-DNJ up to a final concentration of nM, produced viral progeny that were able to infect approximately 20 % of naive MDBK cells. Infected cells incubated with 750-nM NB-DNJ delivered via DCPP
liposomes, however, significantly reduced infectivity of viral progeny (less than 1%
of naive cells were infected). Surprisingly, all infected MDBK cells treated with DCPP liposomes, either with or without encapsulated NB-DNJ, reduced the number of cells infected by viral progeny to approximately 7 %. Therefore, DCPP
liposomes encapsulating only 1 x PBS reduced BVDV infectivity almost 3-fold compared to untreated virus, however, in combination with 750-nM NB-DNJ antiviral effects were increased to over 20-fold greater than controls. NB-DNJ added freely to the medium at the same concentration, 750 nM, had little to no effect. Assays can be further optimized to completely abolish viral infectivity through increasing either liposome or encapsulated NB-DNJ concentrations.
[0123] Although the invention is not limited by its principle of operation, the mechanism, by which viral infectivity is affected by treatment with NB-DNJ
encapsulating DCPP liposomes, can be a combined effect of DCPP lipids integrating into the ER membrane, as well as a targeted delivery of the antiviral agent, NB-DNJ, directly to its site of action leading to enhanced activity and misfolding of viral glycoproteins. As a result, viral progeny treated with these liposomes can have DCPP
lipids present in their viral envelope, in addition to viral glycoproteins that are defective due to the lack of glycan processing in the ER.

ANTIVIRAL EFFECT OF LIPOSOME ENCAPSULATED NB-DNJ
[0124] The antiviral effect of liposome-included NB-DNJ was also tested against the cp BVDV, using the yield reduction assay, a method which takes advantage of the cp strain being able to form plaques on monolayers of infected cells.
[0125] Figure 7 represents the effect of free vs. DC liposome-encapsulated NB-DNJ, on secretion of infectious cp BVDV. MDBK cells infected with cp BVDV were treated for 3 days with either free or DC liposome-NB-DNJ, so that the final NB-DNJ
concentrations ranged between 0 and 500 gM and the total lipid concentration was 100 gM. The supematants were then removed and used to infect fresh MDBK
monolayers in six-well plates. After 3 days the plaques were counted under the microscope (plaque assay) and the results were expressed as a percentage of the number of plaques resulting from infection with no drug supematant (=100%) (x axis). The y axis indicates the free or DC liposome-included NB-DNJ
concentrations used in the plaque assay. The IC50 is indicated at the bottom of the graph.
The results show that NB-DNJ inclusion into the DC liposomes resulted in a 8 fold better inhibition of secretion of infectious BVDV (IC50 of 20 gM, compared to 175 gM
for the free drug). The assay on the cp BVDV can be optimized by incorporating NB-DNJ into DCPP liposomes, which can be more stable in the cell culture medium and hence have an improved antiviral effect.

INCREASED ACTIVITY OF DCPP ENCAPSULATED NB-DNJ MEASURED BY
FREE OLIGOSACCHARIDE ANALYSIS
[0126] Free oligosaccharides (FOS) produced in the presence of free or DCPP
liposomes encapsulating NB-DNJ are characterized and used as a cellular marker for glucosidase inhibition to measure the enhancement of drug activity due to intracellular delivery. FOS have been shown to be generated through the action of a cytosolic peptide:N-glycanase (PNGase) on misfolded glycoproteins exported from the ER for proteasomal degradation via the Sec61-containing channel in the ER
membrane. The identification of glycans present in FOS reveals at what stage protein folding was interrupted. In this experiment, the distribution of glucosylated FOS is used as a measure of glucosidase inhibition by NB-DNJ, where G1ciManõ-FOS and G1c2Manõ-FOS species are a result of glucosidase II inhibition, and G1c3Manõ-FOS a result of glucosidase I inhibition.
[0127] CHO cells were incubated in the presence of free NB-DNJ at the antiviral concentration of 0.5 mM, or with 100 gM of DCPP liposomes encapsulating NB-DNJ
at final concentrations ranging 0 - 75 nM. Cells were left to incubate for 5 days and isolation of FOS was performed using cell homogenates standardized to 500 gg of total protein as described by Mellor et al (2004). Biochem J. 2004 August 1;
381(Pt 3): 861-866. Detection of FOS was performed by normal-phase HPLC following labeling of oligosaccharides by 2-aminobenzamide. The compositions of FOS were further characterized and determined by digestions with glycosidases including endoglycosidase H, jack bean a-mannosidase, and a-glucosidases I and II. The percentage of non-glucosylated and glucosylated oligomannose FOS (represented by Manõ-FOS, G1ciManõ-FOS, G1c2Manõ-FOS, and G1c3Manõ-FOS) was calculated for each sample, and results are represented in Table 2. Results represent the mean ~
S.D. of four separate experiments.
Table 2. Distribution of FOS produced in the presence of free or DCPP liposome-delivered NB-DNJ in CHO cells.
Distribution of total FOS (% S.D.) NB-DNJ Sample G1coManõ G1ciManõ G1c2Manõ G1c3Manõ
untreated 78.8 4.3 16.0 2.4 3.4 0.5 1.8 0.2 0.5 mM free 11.9 1.9 9.5 0.9 31.7 4.5 46.9 4.8 PBS in liposomes 81.2 6.4 15.2 4.8 2.9 1.8 0.7 0.5 0.75 pM in liposomes 70.7 5.4 18.7 3.7 7.7 1.1 2.9 0.9 7.5 pM in liposomes 70.4 4.2 17.7 1.3 8.3 2.1 3.6 1.3 75 pM in liposomes 51.7 4.7 38.6 4.1 5.3 1.8 4.4 2.8 0.75 nM in liposomes 20.4 2.7 66.9 5.1 7.4 3.3 5.3 2.9 7.5 nM in liposomes 12.7 2.6 50.0 4.8 28.6 2.7 8.7 1.7 75 nM in liposomes 13.2 4.1 7.9 2.8 26.6 4.5 52.3 6.2 [0128] CHO cells incubated in the presence of free NB-DNJ at the antiviral concentration of 0.5 mM produced primarily triglucosylated species of FOS, indicating inhibition of glucosidase I at this concentration. This can suggest that inhibition of glucosidase I is responsible for the antiviral effects of NB-DNJ
previously reported for HIV. FOS isolated from liposome-treated samples with increasing 1VB-DNJ concentrations revealed that the distribution of glucosylated FOS
gradually shifted from primarily G1ciMan-FOS to G1c3Man-FOS, with the inhibition of glucosidase II and I, respectively. When delivered via DCPP liposomes, samples treated with a final concentration of 750 nM NB-DNJ demonstrated comparable levels of inhibition of glucosidase I as seen for free incubations at 0.5 mM, suggesting an approximate 600 to 700-fold enhancement of antiviral activity as a result of intracellular delivery.
[0129] Although the invention is not bound by its principle of operation, the mechanism, by which increased NB-DNJ activity is achieved, can be through the direct delivery of this antiviral agent to its site of action (i.e. the ER
lumen). Direct ER delivery can allow the agent to bypass both the plasma and ER membranes, which potentially can act as barriers when NB-DNJ is added freely to the surrounding medium. The increased levels of intracellular delivery observed when NB-DNJ is used for encapsulation may also contribute to an enhancement of activity as a result of higher concentrations of NB-DNJ within the ER lumen compared to other compounds.

INCREASED ACTIVITY OF DCPP ENCAPSULATED NB-DNJ DETERMINED
[0130] In the following example, the activity of NB-DNJ as measured by the inhibition of glycan processing on the HIV envelope protein, gp120, is determined following expression in CHO cells in the presence of free or DCPP liposome-encapsulated NB-DNJ.
[0131] CHO cells expressing a soluble form of HIV gp120 were incubated in the presence of free NB-DNJ with concentrations ranging between 0 - 5 mM, or with liposomes encapsulating NB-DNJ with final concentrations in the medium ranging between 0 and 750 nM. Cells were left to incubate for 5 days before cellular supematant containing the treated gp 120 was collected. To measure the inhibition of glycan processing on gp120 expressed in the presence of NB-DNJ (either free or in liposomes), the binding of the MAb 2G12 was determined by capture ELISA. 2G12 recognizes a cluster of mannose residues on the carbohydrate-rich surface of gp120, and loss of binding in the presence of NB-DNJ results from the retention of glucose on the oligomannose glycans that form the epitope. A loss of binding affinity, however, may also arise from a misfolding of the protein, and to distinguish between these two possibilities, the affinity of all samples for the neutralizing MAb, b12, is also measured. The b12 antibody recognizes a conformationally-sensitive epitope, the CD4 binding site, which does not overlap with that of 2G12. Soluble gp1201ocated in the cellular supematant was captured in ELISA plates using the D7324 antibody (binds the C5 region of gp120) and treated with 10 g/ml of either 2G12 or b12. The binding of both antibodies to NB-DNJ-treated samples was related to that for the untreated gp120, where data are expressed as percent binding and represent the mean S.D. of triplicates from four separate experiments.
[0132] Figure 8 demonstrates a loss of gp120 binding to 2G12 (1.2 0.1 %
binding) following treatment with 0.5-mM NB-DNJ free in the medium, while maintaining % binding to b12. Liposome-mediated delivery with a final concentration of 7.5 nM
NB-DNJ resulted in 9.8 4.8 % binding to 2G12 with no significant effect on b12 binding (96.3 3.2 %), demonstrating a 60,000 to 70,000-fold enhancement of the IC90 as a result of intracellular delivery.

LIPOSOME DELIVERY OF IMMUNOPOTENTIATING PEPTIDES
[0133] The inventors have also discovered that one can increase presentation by major histocompatibility molecule class 1 by contacting an antigen presenting cell with a composition that contains a pH sensitive liposome, such as a liposome comprising DOPE, CHEMS and/or PEG-PE lipids, and an antigen, such as an immunopotentiating peptide, encapsulated in the liposome. Such contacting can be a result of administering the composition to a subject that comprises the antigen presented cell. The administration of the composition to the subject can be used for vaccinating the subject.
[0134] The immunopotenting peptide can be exemplified by a tyrosinase peptide, YMDGTMSQV, which has been found to be presented by a major histocompatibility molecule 1, HLA-A0201, on cells expressing full-length tyrosinase. This is a converted peptide resulted from the tyrosinase peptide, YMNGTMSQV
corresponding to tyrosinase amino acids 369-377 and including the N-linked glycosylation site 6. Although the present invention is not limited by its theory of operation, the converted peptide probably can arise as a result of the deglycosylation in the cytosol by the enzyme peptide: N-glycanase. N-glycanase peptide binds to the transporter associated with antigenic processing (TAP), which transports the peptide into the ER. The encapsulation of the YMDGTMSQV peptide into DCPP liposomes reduces the ER delivery of the peptide by an order of magnitude.

TREATMENT OF HIV-1-INFECTED PBMCs WITH FREE AND LIPOSOME-ENCAPSULATED NB-DNJ
[0135] Clearance of HIV-1 primary isolates from infected human cells by NB-DNJ
was assessed using phytohemagglutinin (PHA)-activated peripheral blood mononuclear cells (PBMCs) as indicator cells and the determination of p24 antigen production as the end point.
[0136] PBMCs from four normal (uninfected) donors were isolated, pooled, and stimulated with PHA (5 g/ml) for 48 h followed by PHA plus interleukin-2 (40 U/ml) for 72 h in RPMI 1640 medium containing 10% heat-inactivated fetal bovine serum (FBS), 100 U of penicillin per ml, 100 g of streptomycin per ml, and 2 mM L-glutamine. All experiments were performed in 96-well microtiter plates. To infect cells, 100 l of PHA-activated PBMCs (5x105/ml) was added to each well, after which an equal volume containing 100 50% tissue culture infective doses (TCID50) of primary isolate stock was added. After an overnight incubation, the cells were washed three times with tissue culture medium, and finally re-suspended in medium containing the appropriate liposome treatment or free NB-DNJ. On day 7, approximately 10 - 30 % of the culture volume containing secreted HIV-1 virions was used to infect naive PBMCs for a second round of infection and treatment (volume transferred was calculated to be the volume necessary for the untreated control of that isolate to infect naive cells at a TCID50 = 100). Rounds of treatment and infection of naive cells were continued over four weeks, and at every time point cellular supernatant containing secreted virions was isolated and used in capture ELISAs to determine p24 concentration (measure of secretion). Additionally, supernatant isolated at each time point throughout treatment was used to infect naive PBMCs (TCID50 = 100) for two weeks with no further treatment, which allowed for the observation of a rebound in viral activity.
[0137] PBMCs infected with 8 primary isolates (listed in Table 3) were incubated in the presence of free NB-DNJ (concentrations ranging 0 - 1 mM) or liposome-encapsulated NB-DNJ (0 - 3.75 M, final lipid concentration of 50 M).
Table 3. List of HIV-1 primary isolates used in assays, including clade identification and tropism.

Primary Primary isolate Clade Tropism isolate Clade Tropism .........................~..................~...........................~......
........................--..................~~............................~
92UG037 A RS 93IN 101 ;C R5 ............................... ~~~~~~~. - 4 92RW021 A RS 97USNG30 ,C R5 ...............................
....................h..............................~

4vvvvvvvvvvvvvvvvvvvvvvvvvvvy\vvvvvvvvvvvvvvvvvvvvv ~vvvvvvvvvvvvvvvvvvvvvvvvvvvvy\vvvvvvvvvvvvvvvvvvvvvvvvvvvvvvv `4vvvvvvvvvvvvvvvvvvvv 4 , ~xxxxxxxxxxxxxxxv~

..........................~......................;\...........................~
................................~ .................... h ..............................~
89.6 B R5/X4 93BR020 F R5/X4 \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \
\ \ \ \ \ \ ~ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \
\ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \
\ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \ \
[0138] Results from PBMCs treated with free 1VB-DNJ confirmed the antiviral concentration against HIV-1 as being 500 M. This was the lowest concentration to clear viral activity over four weeks treatment in all isolates (Figure 9a).
All isolates demonstrated at least a 90 % reduction in viral activity (IC90) suggesting that NB-DNJ is able to target a broad range of HIV-1 effectively.
[0139] The effect of free NB-DNJ on viral secretion can be determined from p24 measurements taken following the first week of treatment. At the highest concentration of NB-DNJ, 1 mM free in medium, most isolates responded with an approximate 30 - 40 % reduction in viral secretion (Figure 9b). Of particular interest are the three clade b isolates, 89.6, JR-FL, and 92HT599, where secretion was reduced by 50 %, and in the case of 89.6, 75 %. 931N101, a clade c isolate, also had a 50 % decrease in secretion.
[0140] The extent of NB-DNJ's antiviral activity on the different primary isolates (drug sensitivity) was estimated from p24 measurements taken following three rounds of treatment. At this point a full curve of p24 secretion was observed for each isolate, and therefore both the IC50 and IC90 could be calculated. Data for the eight isolates tested are shown in Figure 9c, where six isolates are calculated to have an IC50 and IC90 of 400 M and 500 M, respectively, and two isolates, 250 M and 375 M, respectively. The two isolates shown to be the most sensitive to NB-DNJ
treatment, 89.6 and 93BR3020, are also the only isolates included in these experiments known to exhibit dual tropism, whereas the other six isolates are either R5 or X4 (mono) tropic.
[0141] The antiviral activity of NB-DNJ, when encapsulated within DOPE:CHEMS:PEG-PE liposomes, was measured in the same eight isolates to determine enhanced activity with intracellular delivery. Figure 10 shows the level of p24 secretion over four weeks treatment in the eight different isolates tested (graphs A-H) with final concentrations of NB-DNJ ranging 0 - 3.75 M. When virus reduction is compared to results using 500 M free NB-DNJ all isolates demonstrate similar patterns of antiviral activity with final NB-DNJ concentrations ranging 3.75 -37.5 nM, an enhancement of approximately 104- 105-fold. Again, there was variation in the sensitivity of the different isolates to treatment with NB-DNJ
liposomes as new variables exist such as rate of liposome uptake and efficiency of intracellular delivery in addition to drug sensitivity.

TARGETING HIV-1 INFECTED PBMCs WITH sCD4-LIPOSOMES AND
IMMUNOLIPOSOMES
[0142] Increased cellular uptake by targeting liposomes to the gp120/gp4l complex expressed on the surface of HIV-1-infected cells was assessed in nine different primary isolates using a soluble CD4 molecule (sCD4) and several monoclonal antibodies known to bind this complex.
[0143] sCD4-liposome conjugates were created by first chemically reacting the primary amine of sCD4 with N-succinimidyl-S-acetylthiopropionate to create a protected sulfhydryl group, which was then unprotected by deacetylation with hydroxylamine=HCI. Immunoliposomes were prepared by first reducing IgG
molecules with 2-mercaptoethanolamine, an agent that specifically reduces the disulfide bonds in the hinge region between the two heavy chains, creating two half IgG molecules each containing free sulfhydryls. Liposomes were prepared as previously described, however a PE lipid containing a maleimide group (MCC-PE) was incorporated into the bilayer so that the final liposome composition was DOPE:CHEMS:PEG-PE:MCC-PE:Rh-PE (molar ratio 6:4:0.3:0.3:0.1). Unprotected sCD4 molecules and reduced IgG molecules were left to incubate with liposomes overnight at room temperature. Liposomes were purified from free sCD4 or IgG
using size exclusion chromatography.
[0144] Fluorescent-labeled lipids (Rh-PE) were incorporated into the liposome bilayer, and increased endocytosis was calculated from the increase in fluorescence detected in cells following incubation. PBMCs were purified, cultured, and infected in 96-well microtiter plates as previously described. PBMCs were left to incubate with primary isolates (TCIC50 = 100) five days before cells were washed three times with tissue culture medium, and finally resuspended in medium containing the appropriate liposome treatment (final lipid concentration of 50 M). Following a 24 h incubation, PBMCs were isolated, washed twice with 200 1 PBS, and resuspended in 50 l PBS with 1% (vol/vol) Empigen. Fluorescence was measured at ~X = 520 nm and ~,m = 590 nm.
[0145] Six separate monoclonal antibodies, IgGs b6, b12, 2G12, 2F5, X5, and 4E10, were included in the study, however b6 and 4E 10 could not be conjugated to liposomes as they both caused aggregation of lipids.
[0146] Figure 11 represents results obtained using sCD4-liposomes and bl2-, , 2F5-, and X5-immunoliposomes encapsulating 1 mM NB-DNJ expressed as the percentage of liposome uptake in relation to the control. For each infection, there are significant differences in uptake between targeting molecules and the liposome only control (P<0.0001). Liposomes coupled to sCD4 were able to target all nine isolates tested and led to a significant increase in cellular uptake in relation to the liposome only control. 2F5- and b12-immunoliposomes were able to increase liposome uptake in PBMCs infected with 5 and 6 different primary isolates, respectively. 2G12-and X5-immunoliposomes did not target any of the primary isolates tested, and none of the targeting molecules caused significant liposome uptake by non-specific interactions.
[0147] Therefore, sCD4-liposomes may be the best molecule for targeting liposomes to HIV-infected cells. Not only does sCD4 successfully target a broad range of HIV-1 primary isolates, it allows for the increased uptake of liposomes in infected cells via receptor-mediated endocytosis.

TREATMENT OF HIV-1 PBMCs WITH sCD4-LIPOSOME-ENCAPSULATED
NB-DNJ
[0148] Since sCD4-liposomes were shown to have the broadest targeting ability, this liposome preparation was used in p24 secretion assays to compare the antiviral activity of NB-DNJ to that seen with naked liposomes or free delivery. Assays including the same group of eight primary isolates were performed with final NB-DNJ
concentrations ranging 0 - 375 nM. Results are presented in Figure 12, where sCD4-liposomes are shown to provide an additional neutralizing capability, so that all sCD4-liposome treatments, even those containing no NB-DNJ, completely neutralized each primary isolate (graphs A-H).
[0149] Table 4 summarizes data representing the rebound of viral activity following removal of all NB-DNJ treatments. In all cases, once viral activity was reduced to zero (or close to zero), there was no rebound once treatments were removed for two weeks.
Table 4. All average p24 secretion data from HIV-1-infected PBMCs over four weeks treatment with NB-DNJ delivered either free in medium, in liposomes, or in sCD4-liposomes. To measure rebound (-rb) of viral activity, treatments are removed for two weeks before p24 secretion is measured.

% p24 secretion 1VB-DNJ Week Week Week Week Week Week 3- Week 4-Treatment (uM) 1 1-rb 2 2-rb 3 rb Week 4 rb HIV-1 isolate: UG92021 Free 0 100 100 100 100 100 100 100 100 Free 250 90 110 83 101 88 91 77 85 Free 500 85 98 56 62 23 27 8 19 Free 1000 61 72 21 19 5 3 0 0 Liposome 0 92 97 85 97 82 91 81 95 Liposome 0.000375 79 101 76 69 38 41 35 44 Liposome 0.00375 61 80 43 57 11 22 2 9 Liposome 0.0375 44 55 31 45 5 12 2 5 Liposome 0.375 40 47 5 1 0 1 0 0 Liposome 3.75 26 41 0 0 0 0 0 1 sCD4-liposome 0 76 15 2 0 0 1 0 0 sCD4-liposome 0.000375 59 22 3 2 1 0 0 0 sCD4-liposome 0.00375 17 13 1 0 0 1 0 0 sCD4-liposome 0.0375 24 11 0 0 0 0 0 0 sCD4-liposome 0.375 18 16 0 0 0 0 0 0 HIV-1 isolate: UG92046 Free 0 100 100 100 100 100 100 100 100 Free 250 91 99 115 92 91 94 79 89 Free 500 79 75 39 43 10 15 0 0 Free 1000 64 64 15 21 3 1 0 0 Liposome 0 103 104 95 103 102 99 97 96 Liposome 0.000375 78 97 77 82 62 78 59 67 Liposome 0.00375 58 82 57 31 11 23 8 5 Liposome 0.0375 40 64 34 36 7 11 1 1 Liposome 0.375 24 27 1 4 0 0 0 0 Liposome 3.75 22 9 0 1 0 0 0 0 sCD4-liposome 0 74 29 5 3 1 0 0 0 sCD4-liposome 0.000375 43 22 4 0 0 0 0 0 sCD4-liposome 0.00375 16 12 1 1 0 0 0 0 sCD4-liposome 0.0375 21 14 0 0 0 1 0 0 sCD4-liposome 0.375 15 15 0 0 0 0 0 0 HIV-1 isolate: HT92599 Free 0 100 100 100 100 100 100 100 100 Free 250 99 93 84 99 88 95 81 88 Free 500 53 67 19 27 8 17 1 1 Free 1000 55 52 10 15 3 6 0 1 Liposome 0 91 100 82 98 95 102 88 95 Liposome 0.000375 68 94 64 104 66 72 39 68 Liposome 0.00375 54 68 34 18 11 21 1 7 Liposome 0.0375 45 74 19 22 8 11 1 2 Liposome 0.375 38 41 5 2 1 1 0 0 Liposome 3.75 33 11 1 7 1 0 0 0 sCD4-liposome 0 63 10 0 0 1 1 0 0 sCD4-liposome 0.000375 40 12 0 0 0 0 0 0 sCD4-liposome 0.00375 23 2 0 0 0 1 0 1 sCD4-liposome 0.0375 15 1 0 0 0 0 0 1 sCD4-liposome 0.375 16 2 0 0 0 0 0 0 HIV-1 isolate: BR93020 Free 0 100 100 100 100 100 100 100 100 Free 250 89 97 74 77 53 71 55 59 Free 500 60 68 19 3 1 0 1 1 Free 1000 58 44 2 0 0 0 0 0 Liposome 0 102 99 99 89 88 92 91 93 Liposome 0.000375 91 105 75 59 55 59 57 46 Liposome 0.00375 56 88 28 37 19 21 15 9 Liposome 0.0375 48 57 21 12 4 5 3 0 Liposome 0.375 50 39 7 4 1 0 1 0 Liposome 3.75 43 17 1 0 0 1 0 0 sCD4-liposome 0 93 56 13 7 2 0 0 0 sCD4-liposome 0.000375 70 41 9 4 0 0 1 0 sCD4-liposome 0.00375 64 27 6 5 0 0 1 0 sCD4-liposome 0.0375 37 13 2 0 0 0 0 0 sCD4-liposome 0.375 31 9 1 0 0 0 0 0 HIV-1 isolate: 89.6 Free 0 100 100 100 100 100 100 100 100 Free 250 98 81 79 70 54 73 59 66 Free 500 36 35 16 8 2 0 0 0 Free 1000 25 21 1 0 1 0 0 0 Liposome 0 93 89 93 92 91 101 88 92 Liposome 0.000375 85 83 47 33 42 57 31 27 Liposome 0.00375 35 55 12 12 1 0 2 1 Liposome 0.0375 24 32 4 2 0 0 0 0 Liposome 0.375 17 10 0 0 0 0 1 0 Liposome 3.75 8 4 0 0 0 0 0 0 sCD4-liposome 0 54 52 2 2 0 0 1 0 sCD4-liposome 0.000375 13 14 0 0 1 0 0 0 sCD4-liposome 0.00375 13 2 0 0 0 0 0 0 sCD4-liposome 0.0375 7 1 0 0 0 0 0 0 sCD4-liposome 0.375 4 2 0 0 0 0 0 0 HIV-1 isolate: JR-FL

Free 0 100 100 100 100 100 100 100 100 Free 250 107 104 105 101 89 97 93 91 Free 500 64 75 24 9 5 4 0 2 Free 1000 44 56 9 5 1 1 0 0 Liposome 0 92 100 99 97 104 98 96 99 Liposome 0.000375 94 111 98 95 87 93 79 90 Liposome 0.00375 72 77 39 11 11 19 13 19 Liposome 0.0375 50 34 16 14 8 7 5 3 Liposome 0.375 32 16 5 7 1 2 1 0 Liposome 3.75 32 6 0 0 0 0 0 0 sCD4-liposome 0 56 24 4 5 1 0 1 0 sCD4-liposome 0.000375 35 17 1 0 0 0 0 0 sCD4-liposome 0.00375 17 1 0 0 0 0 0 0 sCD4-liposome 0.0375 14 0 0 0 0 0 0 0 sCD4-liposome 0.375 21 0 0 0 0 0 0 0 HIV-1 isolate: 931N101 Free 0 100 100 100 100 100 100 100 100 Free 250 106 96 81 88 78 81 66 72 Free 500 75 88 45 37 7 11 2 0 Free 1000 67 51 17 4 3 1 0 0 Liposome 0 109 102 101 105 96 101 92 95 Liposome 0.000375 96 109 97 101 44 67 36 41 Liposome 0.00375 76 99 53 72 14 18 6 4 Liposome 0.0375 64 57 24 19 5 7 2 1 Liposome 0.375 48 17 6 7 0 0 0 0 Liposome 3.75 48 20 4 1 1 0 0 0 sCD4-liposome 0 86 40 20 21 7 11 0 1 sCD4-liposome 0.000375 61 36 6 9 5 9 1 0 sCD4-liposome 0.00375 40 14 1 3 0 0 0 0 sCD4-liposome 0.0375 20 3 0 0 0 0 0 0 sCD4-liposome 0.375 23 0 0 0 0 0 0 0 HIV-1 isolate: 97USNG30 Free 0 100 100 100 100 100 100 100 100 Free 250 96 112 102 109 100 97 95 101 Free 500 74 82 45 19 8 7 3 1 Free 1000 46 53 9 3 0 0 0 0 Liposome 0 93 99 98 104 97 101 93 95 Liposome 0.000375 83 93 80 79 77 81 62 77 Liposome 0.00375 47 58 32 46 28 32 13 2 Liposome 0.0375 48 36 11 9 9 11 6 3 Liposome 0.375 34 13 1 1 0 0 0 0 Liposome 3.75 20 11 0 0 0 0 0 0 sCD4-liposome 0 73 55 26 15 15 4 1 0 sCD4-liposome 0.000375 41 14 5 9 3 1 0 0 sCD4-liposome 0.00375 20 8 0 0 0 1 0 0 sCD4-liposome 0.0375 17 3 0 0 0 0 0 0 sCD4-liposome 0.375 23 4 1 0 0 0 0 0 INCORPORATION OF PHOSPHATIDYLINOSITOL INTO DOPE:CHEMS
LIPOSOMES
[0150] Phosphatidylinositol (PI) purified from bovine liver cells was incorporated into the previously assayed liposome composition of DOPE:CHEMS:Rh-PE at a final molar concentration of 10-30%. DOPC:CHEMS:Rh-PE liposomes were included in the study as a negative control. Liposomes were prepared as previously described.
MDBK cells were grown to 50 % confluency before media was exchanged and replaced with fresh media containing liposomes with a final lipid concentration of 100 gM. After a 15 m incubation, liposomes were removed and cells were washed twice in PBS. Cells were incubated in fresh media for 0, 1, 2, 5, 24, or 48 hours before cells were fixed in 2.5% paraformaldehyde and visualized under a fluorescent microscope.
Cells were stained with DAPI prior to visual analysis.
[0151] Figure 13 shows representative fluorescent images, taken following a 15 m pulse of each liposome preparation with MDBK cells, at time points 0, 1, 2, 5, 24, and 48 hours. Results demonstrate that liposomes containing PI lipids at final concentrations between 10 and 30% have increased lifetime within the cell, which can mean that lipids delivered via this method are more efficiently retained inside the cell, or alternatively, less efficiently degraded. Fluorescent images taken following DOPE:CHEMS:Rh-PE liposome incubation with MDBK cells show that these lipids are mostly degraded sometime between 5h and 24 h, and completely degraded by h. After 48 h, PI containing liposomes were still very evident within the treated cells, and a more diffuse pattern of Rh-PE is observed. This result can indicate that at this time point most lipids have incorporated into cellular membranes and are no longer concentrated in vesicles (punctate fluorescent pattern).

INCORPORATION OF DOPE:CHEMS LIPOSOMES INTO THE ENVELOPE OF
ER-BUDDING VIRAL PARTICLES
[0152] To investigate whether liposomes are able to fuse with cellular membranes, such as the ER, liposomes containing Rh-PE were used to monitor the uptake into viral particles budding from the ER membrane. MDBK cells persistently infected with a cytopathic BVDV (NADL, MOI=0.01) and naive (uninfected) MDBK are incubated in the presence of DOPE:CHEMS:Rh-PE, DOPE:CHEMS:PI:Rh-PE, or DOPC:CHEMS:Rh-PE liposomes at a final lipid concentration of 50 gM for two days. Following the incubation, media containing liposomes was removed and cells were washed twice in PBS before fresh media was added and cells were incubated a further three days. After three days a sample of supernatant from each set of treated cells (both infected and uninfected) is taken and used for fluorescent measurement using a spectrofluorometer set at ~,X 550 nm and Xem=590 nm. Experiments were carried-out in triplicate, and results represent the average of the three readings.
[0153] Any increase in fluorescence in the supernatant of infected cells compared to the uninfected cells treated with the same liposome preparation is due to the secretion of viral particles containing the Rh-PE lipid in their envelope. Figure 14 shows results obtained using three different lipid compositions, and these data indicate that liposomes containing DOPE (PE) are able to incorporate into the ER membrane, and subsequently into the budding viral envelope. Supernatant from infected cells treated with DOPE liposomes all demonstrated a significant increase in fluorescence compared to uninfected samples. DOPC (PC)-containing liposomes used as a negative control (no ER targeting) demonstrated no difference in fluorescence between infected and uninfected samples. Surprisingly, and in accordance with the data presented in Figure 13, liposomes containing 10-30% PI as part of the lipid composition were able to incorporate into the viral envelopes almost 5 times more efficiently then liposomes without. This indicates that PI is responsible for the increased uptake of liposomes into the ER membrane. This result may have implications for the treatment of viruses that bud from the ER such as BVDV, HCV
and HBV.
[0154] The next step in the development of antiviral therapies using ER-targeting liposomes can be incorporation of antiviral proteins within the lipid bilayer of the liposomes for delivery into the ER membrane. Antiviral proteins can include, but not limited to, viral receptors known to bind viral envelope proteins and/or mutated forms of viral envelope proteins and/or proteins known to interfere with viral envelope interactions.
[0155] The incorporation of antiviral proteins into ER-targeting liposome preparations encapsulating antiviral drugs can provide a combination of three separate antiviral strategies that may act synergistically to reduce viral infectivity:
1- direct delivery of lipids into the ER membrane that when incorporated into viral envelope will reduce viral infectivity, 2- direct delivery of proteins into the ER
membrane that will incorporate into the viral envelope of budding particles and reduce infectivity, and 3- direct delivery of antiviral agents into intracellular compartments.
[0156] Although the foregoing refers to particular preferred embodiments, it will be understood that the present invention is not so limited. It will occur to those of ordinary skill in the art that various modifications may be made to the disclosed embodiments and that such modifications are intended to be within the scope of the present invention.
[0157] All of the publications, patent applications and patents cited in this specification are incorporated herein by reference in their entirety.

Claims (106)

1. A method of treating a viral infection, comprising administering to a host in need thereof a composition comprising (a) a liposome comprising DOPE and CHEMS lipids and (b) one or more compounds encapsulated into the liposome, wherein the viral infection is an ER membrane budding viral infection or a plasma membrane budding viral infection;
wherein the one or more compounds comprise N-butyl deoxynojirimycin (NB-DNJ) and wherein said administering results in delivering of the one or more compounds into an endoplasmic reticulum of a cell, that is infected with a virus causing the infection, and incorporating one or more lipids of the liposome in an endoplasmic reticulum membrane of the cell.
2. The method of claim 1, wherein the infection is an ER membrane budding viral infection.
3. The method of claim 2, wherein the infection is HBV, HCV or BVDV
infection.
4. The method of claim 1, wherein the infection is a plasma membrane budding viral infection.
5. The method of claim 4, wherein the infection is an HIV infection.
6. The method of claim 1, wherein the liposome is a DC liposome.
7. The method of claim 1, wherein the liposome further comprises PEG-PE
lipids.
8. The method of claim 7, wherein the liposome is a DCPP liposome.
9. The method of claim 1, wherein the liposome further comprises PI lipids.
10. The method of claim 9, wherein a molar concentration of the PI lipids in the liposome is from 5 to 50%.
11. The method of claim 10, wherein the molar concentration of the PI lipids in the liposome is from 10 to 30%.
12. The method of claim 1, wherein the one or more compounds further comprise a nucleoside/nucleotide antiviral agent, an immunostimulating/
immunomodulating agent or a combination thereof.
13. The method of claim 12, wherein the nucleoside/nucleotide antiviral agent is ribavirin and the immunostimulating/immunomodulating agent is interferon alpha.
14. The method of claim 1, wherein the one or more compounds further comprise at least one anti-HIV compound.
15. The method of claim 1, wherein the host is a human.
16. A method of treating a viral infection, comprising administering to a host in need thereof a composition comprising (a) a liposome comprising DOPE, CHEMS and PEG-PE lipids and (b) one or more compounds encapsulated into the liposome, wherein the one or more compounds comprise N-butyl deoxynojirimycin (NB-DNJ).
17. The method of claim 16 wherein the virus is a hepatitis virus.
18. The method of claim 17, wherein the virus is hepatitis B virus
19. The method of claim 17, wherein the virus is hepatitis C virus
20. The method of claim 17, wherein the virus is BVDV virus
21. The method of claim 20, wherein the virus is a ncp strain of the BVDV
virus.
22. The method of claim 20, wherein the virus is a cp strain of the BVDV virus
23. The method of claim 18, wherein the one or more compounds further comprise a nucleoside/nucleotide antiviral agent, an immunostimulating/
immunomodulating agent or a combination thereof.
24. The method of claim 23, wherein the nucleoside/nucleotide antiviral agent is ribavirin and the immunostimulating/ immunomodulating agent is interferon.
25. The method of claim 18, wherein the virus is an HIV virus.
26. The method of claim 25, wherein the HIV virus is selected from a group consisting of 92UG037, 92RW021, JR-FL, 92HT599, 89.6, 93IN101, 97USNG30, 92UG021, 92UG046 and 93BR020 primary HIV-1 isolates.
27. The method of claim 25, wherein the one or more compounds further comprise one or more anti-HIV agents.
28. The method of claim 18, wherein the host is a human.
29. A method comprising administering to a host in need thereof a composition comprising (a) a pH
sensitive liposome and (b) an antigen encapsulated inside the liposome, wherein the administering results in increasing antigen presentation by a major histocompatibility molecule class 1 of a antigen presenting cell.
30. The method of claim 29, wherein the antigen is an immunopotentiating peptide.
31. The method of claim 30, wherein the peptide is YMDGTMSQV peptide.
32. The method of claim 29, wherein said liposome comprises DOPE and CHEMS.
33. The method of claim 32, wherein said liposome further comprises PEG-PE
lipids.
34. The method of claim 29, wherein the host is a human.
35. A method of treating an HIV infection comprising administering to a host in need thereof a composition comprising a liposome conjugated with a gp120/gp4l complex targeting moiety.
36. The method of claim 35, wherein the targeting moiety comprises a sCD4 molecule.
37. The method of claim 35, wherein the targeting moiety comprises a monoclonal antibody.
38. The method of claim 37, wherein the antibody is a IgG 2F5 or IgG b12 antibody.
39. The method of claim 35, wherein the HIV infection is caused by an HIV-1 primary isolate selected from the group consisting of 92UG037, 92RW021, JR-FL, 92HT599, 89.6, 931N101, 97USNG30, 92UG021, 92UG046 and 93BR020 primary HIV-1 isolates.
40. The method of claim 35, wherein the liposome comprises DOPE and CHEMS
lipids.
41. The method of claim 40, wherein the liposome further comprises PEG-PE
lipids.
42. The method of claim 40, wherein the liposome further comprises MCC-PE
lipids.
43. The method of claim 40, wherein the liposome further comprises PI lipids.
44. The method of claim 35, wherein the composition further comprises an .alpha.
glucosidase inhibitor encapsulated inside the liposome.
45. The method of claim 44, wherein the inhibitor comprises NB-DNJ.
46. The method of claim 35, wherein the host is a human.
47. A composition comprising a liposome comprising DOPE, CHEMS and PEG-PE lipids and N-butyldeoxynojirimycin (NB-DNJ) encapsulated inside the liposome.
48. The composition of claim 47, wherein the liposome is a DCPP liposome.
49. The composition of claim 47, further comprising a nucleoside/nucleotide antiviral agent, an immunostimulating/immunomodulating agent or a combination thereof.
50. The composition of claim 49, wherein the nucleoside/nucleotide antiviral agent is ribavirin and an immunostimulating/ immunomodulating agent is interferon.
51. The composition of claim 47, further comprising one or more anti-HIV
agents.
52. A composition, comprising a pH sensitive liposome and an antigen encapsulated inside the liposome.
53. The composition of claim 52, wherein the antigen comprises an immunopotentiating peptide.
54. The composition of claim 52, wherein the liposome comprises DOPE and CHEMS lipids.
55. The composition of claim 54, wherein the liposome further comprises PEG-PE

lipids.
56. The composition of claim 54, wherein the liposome further comprises PI
lipids.
57. A composition comprising a liposome conjugated with a gp120/gp41 complex targeting moiety.
58. The composition of claim 57, wherein the targeting moiety comprises a sCD4 molecule.
59. The composition of claim 57, wherein the targeting moiety comprises a monoclonal antibody.
60. The composition of claim 57, wherein the liposome comprises DOPE and CHEMS lipids.
61. The composition of claim 60, wherein the liposome further comprises PEG-PE

lipids.
62. The composition of claim 60, wherein the liposome further comprises MCC-PE lipids.
63. The composition of claim 60, wherein the liposome further comprises PI
lipids.
64. The composition of claim 63, wherein a molar concentration of the PI
lipids in the liposome is from about 10 to about 30%.
65. The composition of claim 57, further comprising an .alpha.-glucosidase inhibitor encapsulated inside the liposome.
66. The composition of claim 65, wherein the .alpha.-glucosidase inhibitor comprises NB-DNJ.
67. A method of treating a viral infection, comprising administering to a host in need thereof a composition comprising (a) a liposome comprising PI lipids and (b) at least one antiviral compound encapsulated into the liposome, wherein the viral infection is an ER membrane virus budding infection;
and wherein said contacting results in delivering of the at least one compound into an endoplasmic reticulum of a cell, that is infected with a virus causing the infection, and incorporating one or more lipids of the liposome in an endoplasmic reticulum membrane of the cell.
68. The method of claim 67, wherein the liposome further comprises DOPE and CHEMS lipids.
69. The method of claim 67, wherein a molar concentration of the PI lipids in the liposome is from 5 to 50%.
70. The method of claim 69, wherein the molar concentration of the PI lipids in the liposome is from 10 to 30%.
71. The method of claim 67, wherein the at least one antiviral compound comprises the .alpha.-glucosidase inhibitor.
72. The method of claim 71, wherein the .alpha.-glucosidase inhibitor comprises NB-DNJ.
73. The method of claim 67, wherein the host is a human.
74. A composition comprising a liposome comprising PI lipids and at least one antiviral compound encapsulated inside the liposome.
75. The composition of claim 74, wherein the liposome further comprises DOPE
and CHEMS lipids.
76. The composition of claim 74, wherein a molar concentration of the PI
lipids in the liposome is from 5 to 50%.
77. The composition of claim 76, the molar concentration of the PI lipids in the liposome is from 10 to 30%.
78. The composition of claim 74, wherein the at least one antiviral compound comprises an a-glucosidase inhibitor.
79. The composition of claim 78, wherein the .alpha.-glucosidase inhibitor comprises NB-DNJ.
80. A method of treating a viral infection comprising administering to a host in need thereof a composition comprising (a) a liposome comprising PI lipids and (b) at least one antiviral protein intercalated into a lipid layer of the liposome, wherein said contacting results in incorporating one or more lipids of the liposome in an endoplasmic reticulum membrane of a cell, that is infected with a virus causing the infection.
81. The method of claim 80, wherein the liposome further comprises DOPE and CHEMS lipids.
82. The method of claim 81, wherein the liposome further comprises PEG-PE
lipids.
83. The method of claim 80, wherein a molar concentration of the PI lipids in the liposome is from 5 to 50%.
84. The method of claim 83, wherein the molar concentration of the PI lipids is from 10 to 30%.
85. The method of claim 80, wherein the composition further comprises (c) at least one antiviral compound encapsulated into the liposome and wherein said contacting results in delivering of the at least one compound into an endoplasmic reticulum of the cell.
86. The method of claim 85, wherein the at least one antiviral compound comprises an .alpha.-glucosidase inhibitor.
87. The method of claim 86, wherein the .alpha.-glucosidase inhibitor comprises NB-DNJ.
88. The method of claim 80, wherein the at least one antiviral protein is selected from the group consisting of viral receptors, mutated forms of viral envelope proteins and proteins interfering with viral envelope interaction.
89. The method of claim 80, wherein the viral infection is an ER membrane budding viral infection.
90. The method of claim 89, wherein the ER budding viral infection is HBV, HCV or BVDV infection.
91. The method of claim 80, wherein the infection is a plasma membrane budding infection.
92. The method of claim 91, wherein the plasma membrane budding infection is an HIV infection.
93. The method of claim 80, wherein the subject is a human.
94. A composition comprising (a) a liposome comprising PI lipids and (b) at least one antiviral protein intercalated into a lipid layer of the liposome.
95. The composition of claim 94, wherein the liposome further comprises DOPE
and CHEMS lipids.
96. The composition of claim 95, wherein the liposome further comprises PEG-PE

lipids.
97. The composition of claim 94, wherein a molar concentration of the PI
lipids in the liposome is from 5 to 50%.
98. The composition of claim 97, wherein the molar concentration of the PI
lipids is from 10 to 30%.
99. The composition of claim 94, wherein the composition further comprises (c) at least one antiviral compound encapsulated into the liposome.
100. The composition of claim 99, wherein the at least one antiviral compound comprises an .alpha.-glucosidase inhibitor.
101. The composition of claim 100, wherein the .alpha.-glucosidase inhibitor comprises NB-DNJ.
102. The composition of claim 94, wherein the at least one antiviral protein is selected from the group consisting of viral receptors, mutated forms of viral envelope proteins and proteins interfering with viral envelope interaction.
103. A composition comprising a liposome comprising PI lipids and at least one therapeutic agent encapsulated inside the liposome.
104. A method of treating or preventing a physiological condition comprising administering to a subject in need thereof a composition comprising a liposome comprising PI lipids and at least one therapeutic agent encapsulated inside the liposome.
105. A composition comprising a liposome comprising PI lipids and at least one protein intercalated into a lipid bilayer of the liposome.
106. A method of treating or preventing a physiological condition comprising administering to a subject in need thereof a composition comprising a liposome comprising PI lipids and at least one protein intercalated into a lipid bilayer of the liposome.
CA002659858A 2006-08-02 2007-08-02 Liposome treatment of viral infections Abandoned CA2659858A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US83479706P 2006-08-02 2006-08-02
US60/834,797 2006-08-02
US84634406P 2006-09-22 2006-09-22
US60/846,344 2006-09-22
PCT/US2007/075080 WO2008088581A2 (en) 2006-08-02 2007-08-02 Liposome treatment of viral infections

Publications (1)

Publication Number Publication Date
CA2659858A1 true CA2659858A1 (en) 2008-07-24

Family

ID=39636531

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002659858A Abandoned CA2659858A1 (en) 2006-08-02 2007-08-02 Liposome treatment of viral infections

Country Status (7)

Country Link
US (2) US20080138351A1 (en)
EP (3) EP2049126A2 (en)
JP (1) JP2009545621A (en)
KR (1) KR20090040906A (en)
CN (1) CN102727437A (en)
CA (1) CA2659858A1 (en)
WO (1) WO2008088581A2 (en)

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2049126A2 (en) * 2006-08-02 2009-04-22 United Therapeutics Corporation Liposome treatment of viral infections
US20080131398A1 (en) * 2006-08-21 2008-06-05 United Therapeutics Corporation Combination therapy for treatment of viral infections
CN102046151A (en) * 2008-03-26 2011-05-04 牛津大学 Endoplasmic reticulum targeting liposomes
US8911711B2 (en) * 2008-09-30 2014-12-16 The Invention Science Fund I, Llc Method, device, and system to control pH in pulmonary tissue of a subject
WO2010096764A1 (en) 2009-02-23 2010-08-26 United Therapeutics Corporation Iminosugars and methods of treating viral diseases
ES2562635T3 (en) * 2009-02-24 2016-03-07 United Therapeutics Corporation Iminoazúcares and methods of treatment of infections caused by arenavirus
CA2757026A1 (en) * 2009-03-27 2010-09-30 The Chancellor, Masters And Scholars Of The University Of Oxford Cholesterol level lowering liposomes
CN102639133B (en) 2009-06-12 2015-03-11 联合治疗公司 Iminosugars and methods of treating bunyaviral and togaviral diseases
KR101435953B1 (en) * 2009-07-17 2014-09-02 한림대학교 산학협력단 Immunostimulatory Compositions Comprising Liposome―Encapsulated Oligonucleotides and Epitopes
DK2473482T3 (en) * 2009-09-04 2014-05-12 United Therapeutics Corp Methods of treating orthomyxoviral infections
CN105748476A (en) * 2009-09-04 2016-07-13 联合治疗公司 Iminosugars And Methods Of Treating Filoviral Diseases
JP5575246B2 (en) * 2009-09-04 2014-08-20 ユナイテッド セラピューティクス コーポレイション How to treat poxvirus infection
JP2013512945A (en) 2009-12-07 2013-04-18 ザ チャンセラー,マスターズ アンド スカラーズ オブ ザ ユニバーシティ オブ オックスフォード N-substituted deoxynojirimycin compounds for use in the inhibition of osteoclastogenesis and / or osteoclast activation
JP2013518814A (en) 2010-02-02 2013-05-23 ザ チャンセラー,マスターズ アンド スカラーズ オブ ザ ユニバーシティ オブ オックスフォード Synthesis of DGJNAc from D-glucuronolactone and use to inhibit α-N-acetylgalactosaminidase
WO2012020108A2 (en) * 2010-08-11 2012-02-16 Cormus Srl Multimeric inhibitors of viral fusion and uses thereof
KR20150035767A (en) 2012-06-06 2015-04-07 유니터 바이롤로지, 엘엘씨 Novel iminosugars and their applications
WO2014169207A1 (en) * 2013-04-11 2014-10-16 Board Of Regents Of The University Of Nebraska Compositions and methods for the delivery of therapeutics
AU2014318488B2 (en) 2013-09-16 2019-03-07 Emergent Virology Llc Deoxynojirimycin derivatives and methods of their using
EP3215499B1 (en) 2014-11-05 2021-01-20 Emergent Virology LLC Iminosugars useful for the treatment of viral diseases
CN107028887A (en) * 2017-03-28 2017-08-11 江苏孟德尔基因科技有限公司 Liposome is used for the purposes for treating chronic viral hepatitis type B

Family Cites Families (84)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2249552A1 (en) * 1971-10-12 1973-05-30 Inchema S A PROCESS FOR THE INCAPSULATION OF IN PARTICULAR WATER-SOLUBLE COMPOUNDS
NO154918C (en) 1977-08-27 1987-01-14 Bayer Ag ANALOGUE PROCEDURE FOR THE PREPARATION OF THERAPEUTIC ACTIVE DERIVATIVES OF 3,4,5-TRIHYDROXYPIPERIDINE.
DE2834122A1 (en) * 1978-08-03 1980-02-14 Bayer Ag METHOD FOR PRODUCING 6-AMINO-6-DESOXY-L-SORBOSE
DE2853573A1 (en) 1978-12-12 1980-07-03 Bayer Ag PREPARATION OF N-SUBSTITUTED DERIVATIVES OF L-DESOXYNOJIRIMYCIN
DE3038901A1 (en) 1980-10-15 1982-05-06 Bayer Ag, 5090 Leverkusen METHOD FOR PRODUCING N-SUBSTITUTED DERIVATIVES OF 1-DESOXYNOJIRIMYCIN
US4690486A (en) * 1985-04-29 1987-09-01 Texas Instruments Incorporated Four position interlacing apparatus
US4837237A (en) * 1985-07-09 1989-06-06 Fred Hutchinson Cancer Research Center Therapy using glucosidase processing inhibitors
DE3543999A1 (en) 1985-12-13 1987-06-19 Bayer Ag HIGH PURITY ACARBOSE
EP0252962B1 (en) * 1985-12-23 1995-08-23 Fred Hutchinson Cancer Research Center Regulating retroviral replication, infection, and pathogenesis
US4925796A (en) * 1986-03-07 1990-05-15 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
DE3611841A1 (en) 1986-04-09 1987-10-15 Bayer Ag METHOD FOR PRODUCING 1-DESOXYNOJIRIMYCIN AND ITS N-DERIVATIVES
US5229376A (en) * 1986-09-25 1993-07-20 The United States Of America As Represented By The Secretary Of The Army Encapsulated plant-derived phosphatidylinositol (PI) compositions for the prevention of mitogenically induced cell proliferation
US4792558A (en) * 1987-05-29 1988-12-20 Merrell Dow Pharmaceuticals Inc. Castanospermine for inhibiting tumor metastasis
US5004746A (en) * 1987-09-29 1991-04-02 Merrell Dow Pharmaceuticals Inc. Anti-retroviral castanospermine esters
US4849430A (en) 1988-03-09 1989-07-18 Monsanto Company Method of inhibiting virus
EP0324328B1 (en) * 1987-12-21 1997-10-22 Monsanto Company Use of the N-n-butyl derivative of desoxynojirimycin in the manufacture of a medicament inhibiting HIV-Virus
US4861892A (en) 1988-02-12 1989-08-29 G. D. Searle & Co. Method for synthesis of deoxymannojirimycin
US5691346A (en) * 1988-08-10 1997-11-25 The Australian National University Castanospermine as an anti-inflammatory and immunosuppressant agent
US5837709A (en) * 1988-08-10 1998-11-17 The Australian National University Use of castanospermine as an anti-inflammatory and immunosupressant agent
US4910310A (en) 1988-10-03 1990-03-20 G. D. Searle & Co. Synthesis of N-substituted 1,5-dideoxy-1,5-imino-L-fucitol derivatives
US4876268A (en) * 1988-11-03 1989-10-24 G. D. Searle & Co. Antiviral compounds and use thereof
US5310745A (en) * 1988-11-03 1994-05-10 G. D. Searle & Co. Antiviral compounds
US4952585A (en) * 1988-12-15 1990-08-28 Merrell Dow Pharmaceuticals Inc. Castanospermine esters in the inhibition of tumor metastasis
US4894388A (en) * 1988-12-22 1990-01-16 Monsanto Company Glycosidase inhibitors and use thereof
US5009376A (en) * 1989-04-26 1991-04-23 Usui Kokusai Sangyo Kaisha Ltd. Seizure-fixing device of pipe by clamp member
US5011929A (en) 1989-05-15 1991-04-30 Monsanto Company Synthesis of mannojirimycin derivatives
US5017704A (en) 1989-06-27 1991-05-21 Monsanto Company Fucosidase inhibitor
US5100797A (en) 1989-06-27 1992-03-31 Monsanto Company Fucosidase inhibitors
US5043273A (en) 1989-08-17 1991-08-27 Monsanto Company Phosphorylated glycosidase inhibitor prodrugs
US4994572A (en) 1989-10-12 1991-02-19 Monsanto Company Synthesis of nojirimycin derivatives
US5214050A (en) * 1989-10-17 1993-05-25 Merrell Dow Pharmaceuticals Inc. Esters of castanospermine in the treatment of cerebral malaria
US4996329A (en) 1989-10-20 1991-02-26 Monsanto Company Derivatives of 1,4-dideoxy-1,4-imino-D-mannitol and a process for their preparation
US5013842A (en) 1990-01-22 1991-05-07 Monsanto Company Synthesis of chiral pyrrolidine and piperidine glycosidase inhibitors
US5200523A (en) 1990-10-10 1993-04-06 Monsanto Company Synthesis of nojirimycin derivatives
GB9027433D0 (en) * 1990-12-18 1991-02-06 Merrell Dow Pharma Anti-herpes castanospermine esters
US5401645A (en) 1992-03-16 1995-03-28 Monsanto Company Process for producing n-substituted polyhydroxy nitrogen-containing heterocycles utilizing acetobacteraceae and corynebacterium
AU668934B2 (en) * 1992-04-10 1996-05-23 Hisamitsu Pharmaceutical Co., Inc. Liposome composition
US5286877A (en) 1993-02-01 1994-02-15 G. D. Searle & Co. Synthesis of 1,4-dideoxy-1,4-imino-L-arabinitol
US5399567A (en) * 1993-05-13 1995-03-21 Monsanto Company Method of treating cholera
US6291657B1 (en) 1993-05-13 2001-09-18 Monsanto Company Deoxygalactonojirimycin derivatives
US5750648A (en) * 1993-08-20 1998-05-12 G.D. Searle & Co. Retroviral protease inhibitors and combinations thereof
WO1995022975A1 (en) 1994-02-25 1995-08-31 G.D. Searle & Co. Use of 1-deoxynojirimycin and its derivatives for treating mammals infected with respiratory syncytial virus
US6773719B2 (en) * 1994-03-04 2004-08-10 Esperion Luv Development, Inc. Liposomal compositions, and methods of using liposomal compositions to treat dislipidemias
US5709865A (en) * 1994-11-10 1998-01-20 Biostar Inc. Immunogenic composition against Bovine Viral Diarrhea Virus II glycoprotein 53 (BVDV-II gp53)
US5591448A (en) * 1994-11-21 1997-01-07 Tepic; Slobodan Anti-viral therapeutic composition
CA2218515C (en) * 1995-04-19 2008-10-07 Polymun Scientific Immunbiologische Forschung Gmbh Monoclonal antibodies against hiv-1 and vaccines made thereof
US6387365B1 (en) 1995-05-19 2002-05-14 Schering Corporation Combination therapy for chronic hepatitis C infection
US6041252A (en) * 1995-06-07 2000-03-21 Ichor Medical Systems Inc. Drug delivery system and method
US5908621A (en) * 1995-11-02 1999-06-01 Schering Corporation Polyethylene glycol modified interferon therapy
DE19623950A1 (en) * 1996-06-15 1997-12-18 Boehringer Ingelheim Kg Pharmaceutical preparation in the form of liposomes
US5908867A (en) * 1996-07-18 1999-06-01 Henry; James P. Reduction of hair growth
ATE371463T1 (en) * 1997-02-05 2007-09-15 Jean-Claude Bystryn PH-SENSITIVE LIPOSOMES AND OTHER TYPES OF ENCAPSULED VACCINES CONTAINING IMMUNOMODULATORS AND METHODS OF PRODUCTION AND USE THEREOF
US6172046B1 (en) 1997-09-21 2001-01-09 Schering Corporation Combination therapy for eradicating detectable HCV-RNA in patients having chronic Hepatitis C infection
US6472373B1 (en) 1997-09-21 2002-10-29 Schering Corporation Combination therapy for eradicating detectable HCV-RNA in antiviral treatment naive patients having chronic hepatitis C infection
EP1030839B1 (en) 1997-11-10 2004-02-04 G.D. SEARLE &amp; CO. Use of alkylated iminosugars to treat multidrug resistance
JP2001525367A (en) * 1997-12-11 2001-12-11 ザ・チャンセラー・マスターズ・アンド・スカラーズ・オブ・ザ・ユニバーシティ・オブ・オックスフォード Inhibition of membrane-associated virus replication
US6465488B1 (en) * 1997-12-11 2002-10-15 Chancellor, Masters & Scholars Of The University Of Oxford Inhibition of glycolipid biosynthesis
ID27290A (en) 1998-02-12 2001-03-22 Searle & Co USE OF 1,5-DIDEOKSI-1,5-IMINO-D-GLUSITOL-N-SUBSTITUTED N-COMPOUND IN TREATMENT OF HEPATITIC VIRUS INFECTION
US6689759B1 (en) 1998-02-12 2004-02-10 G. D. Searle & Co. Methods of Treating hepatitis virus infections with N-substituted-1,5-dideoxy-1,5-imino-d-glucitol compounds in combination therapy
US20020151683A1 (en) * 1998-03-30 2002-10-17 Mogam Biotechnology Research Institute Liposomes comprising peptide antigens derived from X protein of hepatitis B virus
US6274597B1 (en) * 1998-06-01 2001-08-14 Mount Sinai School Of Medicine Of New York University Method of enhancing lysosomal α-Galactosidase A
US6824768B2 (en) * 1998-12-18 2004-11-30 Schering Corporation Ribavirin-pegylated interferon alfa induction HCV combination therapy
AU3858600A (en) * 1999-02-12 2001-02-19 G.D. Searle & Co. Glucamine compounds for treating hepatitis virus infections
EP1165080A2 (en) 1999-02-12 2002-01-02 G.D. SEARLE &amp; CO. Use of substituted-1,5-dideoxy-1,5-imino-d-glucitol compounds for treating hepatitis virus infections
US6852334B1 (en) * 1999-04-20 2005-02-08 The University Of British Columbia Cationic peg-lipids and methods of use
JP2003506406A (en) 1999-08-10 2003-02-18 ザ・チャンセラー・マスターズ・アンド・スカラーズ・オブ・ザ・ユニバーシティ・オブ・オックスフォード Long-chain N-alkyl compounds and oxa derivatives thereof
US7256005B2 (en) 1999-08-10 2007-08-14 The Chancellor, Masters And Scholars Of The University Of Oxford Methods for identifying iminosugar derivatives that inhibit HCV p7 ion channel activity
EP1250129A2 (en) * 2000-01-20 2002-10-23 Washington University METHODS TO TREAT $g(a)-1-ANTITRYPSIN DEFICIENCY
AU2001234596A1 (en) 2000-01-28 2001-08-07 Timothy M Block Use of castanospermine and substituted-castanospermine compounds for treating hepatitis virus infections
AU2001278336B2 (en) * 2000-07-31 2007-01-18 Ottawa Heart Institute Research Corporation Charged phospholipid compositions and methods for their use
CN100544718C (en) * 2001-04-20 2009-09-30 维奥恩药品公司 The purposes of chemical compound in the preparation antiviral agent
AU2002348971A1 (en) * 2001-10-12 2003-04-28 Stefana M. Petrescu Ph-sensitive liposomes for targeted drug delivery
AU2002359327A1 (en) * 2001-10-30 2003-05-12 Thomas Jefferson University Method of treating viral infections
JP4555569B2 (en) * 2001-11-13 2010-10-06 セレーター ファーマシューティカルズ, インコーポレイテッド Lipid carrier composition having enhanced blood stability
WO2003075890A1 (en) * 2002-03-05 2003-09-18 Transave, Inc. Methods for entrapment of bioactive agent in a liposome or lipid complex
EP1631669A2 (en) * 2003-04-09 2006-03-08 Biodelivery Sciences International, Inc. Cochleate compositions directed against expression of proteins
EA200700718A1 (en) * 2004-10-06 2007-12-28 Мидженикс Инк. COMBINED ANTI-VIRAL COMPOSITIONS CONTAINING KASTANOSPERMIN AND METHODS OF THEIR APPLICATION
JP5057362B2 (en) * 2004-11-02 2012-10-24 国立大学法人 東京医科歯科大学 Liposomes and methods for injecting substances into cells using the same
JP2008530124A (en) * 2005-02-09 2008-08-07 ミジェニックス インコーポレイテッド Compositions and methods for treating or preventing flavivirus infections
JP5241709B2 (en) * 2006-05-24 2013-07-17 ユナイテッド セラピューティクス コーポレーション Deoxynojirimycin and D-arabinitol analogs and methods of use
EP2049126A2 (en) * 2006-08-02 2009-04-22 United Therapeutics Corporation Liposome treatment of viral infections
US20080131398A1 (en) * 2006-08-21 2008-06-05 United Therapeutics Corporation Combination therapy for treatment of viral infections
CN102046151A (en) * 2008-03-26 2011-05-04 牛津大学 Endoplasmic reticulum targeting liposomes
CA2757026A1 (en) * 2009-03-27 2010-09-30 The Chancellor, Masters And Scholars Of The University Of Oxford Cholesterol level lowering liposomes

Also Published As

Publication number Publication date
US20110182982A1 (en) 2011-07-28
WO2008088581A3 (en) 2009-02-26
EP2399587A1 (en) 2011-12-28
KR20090040906A (en) 2009-04-27
CN102727437A (en) 2012-10-17
EP2356990A3 (en) 2011-10-19
EP2049126A2 (en) 2009-04-22
US20080138351A1 (en) 2008-06-12
JP2009545621A (en) 2009-12-24
WO2008088581A2 (en) 2008-07-24
EP2356990A2 (en) 2011-08-17

Similar Documents

Publication Publication Date Title
US20080138351A1 (en) Liposome treatment of viral infections
US8703744B2 (en) Cholesterol level lowering liposomes
US20120237592A1 (en) Endoplasmic reticulum targeting liposomes
US20070161544A1 (en) Selective targeting agents for mitcochondria
AU8285998A (en) Targeted liposomal constructs for diagnostic and therapeutic uses
JP2009512716A (en) Small molecule inhibitors of HIV-1 capsid construction
US9006186B2 (en) Selective targeting agents for mitochondria
WO2006077427A2 (en) Antiviral drug combinations
Szebeni et al. Complement activation, immunogenicity, and immune suppression as potential side effects of liposomes
Désormeaux et al. Liposomes as drug delivery system: a strategic approach for the treatment of HIV infection
US8791075B2 (en) Methods and compositions for inhibition of BCL6 repression
AU768685B2 (en) Methods and formulations for targeting infectious agents bearing host cell proteins
JPH06504429A (en) Inhibition of viral replication
KR20230158020A (en) Compounds for use in the treatment of enveloped viral infections
CN101557813A (en) Liposome treatment of viral infections
RU2316320C1 (en) Anti-viral agent
US20180177724A1 (en) Site-targeted nano-liposomal nitroglycerin therapeutics
Düzgünes Treatment of human immunodeficiency virus, Mycobacterium avium, and Mycobacterium tuberculosis infections by liposome-encapsulated drugs
EP3636255A1 (en) Use of deoxysphingolipids as anti-toxins and anti-virals
WO2003105677A2 (en) Methods for treating viral diseases using modulators of amyloidogenic peptide aggregation
Weissig et al. 11 Mitochondria-Targeted Drug Delivery
Sau The molecular mechanisms underlying the in vitro immunostimulatory and toxic effects of the antifungal drug, amphotericin B: Role of Toll-like receptor 2 and CD14

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued

Effective date: 20141205